US10253100B2 - Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance - Google Patents

Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance Download PDF

Info

Publication number
US10253100B2
US10253100B2 US14/347,187 US201214347187A US10253100B2 US 10253100 B2 US10253100 B2 US 10253100B2 US 201214347187 A US201214347187 A US 201214347187A US 10253100 B2 US10253100 B2 US 10253100B2
Authority
US
United States
Prior art keywords
eu434y
eu436v
eu252y
eu440e
eu254t
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US14/347,187
Other languages
English (en)
Other versions
US20140363428A1 (en
Inventor
Tomoyuki Igawa
Atsuhiko Maeda
Futa Mimoto
Taichi Kuramochi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chugai Pharmaceutical Co Ltd
Original Assignee
Chugai Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/JP2012/058603 external-priority patent/WO2012133782A1/ja
Application filed by Chugai Pharmaceutical Co Ltd filed Critical Chugai Pharmaceutical Co Ltd
Assigned to CHUGAI SEIYAKU KABUSHIKI KAISHA reassignment CHUGAI SEIYAKU KABUSHIKI KAISHA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KURAMOCHI, TAICHI, IGAWA, TOMOYUKI, MAEDA, ATSUHIKO, MIMOTO, FUTA
Publication of US20140363428A1 publication Critical patent/US20140363428A1/en
Application granted granted Critical
Publication of US10253100B2 publication Critical patent/US10253100B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2812Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/526CH3 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates to: a modified FcRn-binding domain having an enhanced affinity for the Fc Receptor neonatal (FcRn) at neutral pH; an antigen-binding molecule comprising said FcRn-binding domain, which has low immunogenicity, high stability and form only a few aggregates; a modified antigen-binding molecule having an increased FcRn-binding activity at neutral or acidic pH without an increased binding activity at neutral pH for a pre-existing anti-drug antibody; use of the antigen-binding molecules for improving antigen-binding molecule-mediated antigen uptake into cells; use of the antigen-binding molecules for reducing the plasma concentration of a specific antigen; use of the modified FcRn-binding domain for increasing the total number of antigens to which a single antigen-binding molecule can bind before its degradation; use of the modified FcRn-binding domain for improving pharmacokinetics of an antigen-binding molecule; methods for decreasing the binding activity for
  • a conventional antibody targeting a soluble antigen binds the antigen in the plasma of the patient after injection and then stably persists in the form of an antibody-antigen complex until degradation. While a typical antibody has generally a long half-life (1-3 weeks), an antigen has a relatively short half-life of less than one day. An antigen in complex with an antibody therefore has a significantly longer half-life than the antigen alone. Consequently, the antigen concentration tends to increase after the injection of a conventional antibody. Such cases have been reported for antibodies targeting various soluble antigens, such as IL-6 (J Immunotoxicol. 2005, 3, 131-9.
  • NPL 1 beta amyloid
  • MAbs. 2010 September-October; 2(5):576-88 NPL 2
  • MCP-1 ARTHRITIS & RHEUMATISM 2006, 54, 2387-92
  • NPL 4 hepcidin
  • sIL-6 receptor Blood. 2008 Nov. 15; 112(10):3959-64.
  • Reports have described an approximately 10 to 1000-fold increase (depending of the antigen) of total plasma antigen concentration from the baseline upon antibody administration.
  • FcRn neonatal Fc receptor for IgG
  • PTL 1 the neonatal Fc receptor for IgG
  • the FcRn is a protein found in the membrane of many cells.
  • An antibody with increased binding activity to FcRn at neutral pH will bind FcRn on the cell surface, whereby the receptor with the antibody is internalized into the cells in a vesicle.
  • a pH-dependent antigen binding antibody having increased binding activity to FcRn at neutral pH can be used to remove an antigen from plasma and decrease its concentration in plasma.
  • An objective of the present invention is to provide a modified FcRn-binding domain which has an enhanced affinity for the FcRn at neutral pH; an antigen-binding molecule comprising said FcRn-binding domain, wherein said antigen-binding molecule has low immunogenicity, high stability and forms only few aggregates; a modified antigen-binding molecule having an increased FcRn-binding activity at neutral or acidic pH without an increased binding activity at neutral pH for a pre-existing anti-drug antibody; use of the antigen-binding molecules for improving antigen-binding molecule-mediated antigen uptake into cells; use of the antigen-binding molecule for reducing the plasma concentration of a specific antigen; use of the modified FcRn-binding domain for increasing the total number of antigens to which a single antigen-binding molecule can bind before its degradation; use of the modified FcRn-binding domain for improving pharmacokinetics of an antigen
  • the present inventors conducted dedicated studies on modified FcRn-binding domains which have an enhanced affinity for FcRn at neutral pH and on antigen-binding molecules comprising said FcRn-binding domain which have low immunogenicity, high stability and form only few aggregates. As a result, the present inventors discovered that substitutions at specific positions of the FcRn-binding domain increases the affinity for the FcRn at neutral pH without substantially increasing the immunogenicity, without substantially decreasing the stability and/or without substantially increasing the ratio of high molecular weight species.
  • the present inventors conducted dedicated studies on modified FcRn-binding domains with an enhanced affinity for FcRn at neutral pH or acidic pH but without a significantly increased binding activity for a pre-existing anti-drug antibody and on antigen-binding molecules comprising such an FcRn-binding domain.
  • substitutions at specific positions of the FcRn-binding domain decrease the affinity for a pre-existing anti-drug antibody at neutral pH without substantially decreasing the FcRn-binding activity.
  • the present invention relates to:
  • An antigen-binding molecule comprising a modified FcRn-binding domain, wherein the modified FcRn-binding domain comprises an amino acid substitution at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, wherein the numbers indicate the position of the substitution according to the EU numbering.
  • the modified FcRn-binding domain comprises at position EU238 an aspartic acid, at position EU250 a valine, at position EU252 a tyrosine, at position EU254 a threonine, at position EU255 a leucine, at position EU256 a glutamic acid, at position EU258 an aspartic acid or an isoleucine, at position EU286 a glutamic acid, at position EU307 a glutamine, at position EU308 a proline, at position EU309 a glutamic acid, at position EU311 an alanine or a histidine, at position EU315 an aspartic acid, at position EU428 an isoleucine, at position EU433 an alanine, a lysine, a proline, an arginine, or a serine, at position EU434 a tyrosine, or a tryp
  • the antigen-binding molecule according to [8], wherein the FcRn-binding domain comprises: a) at position EU252 a tyrosine, at position EU286 a glutamic acid, at position EU307 a glutamine, at position EU311 an alanine, at position EU434 a tyrosine, and at position EU436 a valine; or b) at position EU250 a valine, at position EU252 a tyrosine, at position EU307 a glutamine, at position EU308 proline, at position EU311 an alanine, at position EU434 a tyrosine, and at position EU436 a valine; or c) at position EU250 a valine, at position EU252 a tyrosine, at position EU286 a glutamic acid, at position EU307 a glutamine, at position EU308 proline, at position EU311 an alanine, at position EU434 a valine
  • the antigen-binding molecule according to [10], wherein the FcRn-binding domain comprises: a) at position EU307 a glutamine, at position EU311 a histidine and at position EU434 a tyrosine; or b) at position EU307 a glutamine, at position EU309 a glutamic acid, at position EU311 an alanine and at position EU434 a tyrosine; or c) at position EU307 a glutamine, at position EU309 a glutamic acid, at position EU311 an histidine and at position EU434 a tyrosine; or d) at position EU250 a valine; at position EU252 a tyrosine, at position EU434 a tyrosine and at position EU436 a valine.
  • antigen-binding molecule according to any one of [1] to [11], wherein the ratio of high molecular weight species is less than 2%.
  • antigen-binding molecule according to any one of [1] to [12], wherein antigen-binding molecule comprises an antigen-binding domain having a) a lower binding activity for the antigen at pH 5.5-6.5 than at pH 7-8 or b) a “calcium concentration-dependent binding” activity for the antigen.
  • [18] The antigen-binding molecule according to any one of [1] to [17], wherein a) at the position EU257 of the FcRn-binding domain the amino acid is not an amino acid selected from the group consisting of alanine, valine, isoleucine, leucine, and threonine, and/or b) at the position EU252 of the FcRn-binding domain the amino acid is not tryptophan.
  • the antigen-binding molecule according to any one of [1] to [18], wherein the antigen-binding molecule has a binding activity for a pre-existing anti-drug antibody that is not significantly increased as compared to the binding affinity of a control antibody comprising an intact FcRn-binding domain.
  • the FcRn binding domain further comprises an amino acid substitution at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the FcRn binding domain comprises one or more amino acid substitutions selected from the group consisting of at position EU387 an arginine, at position EU422 a glutamic acid, an arginine, or a serine, an aspartic acid, a lysine, a threonine or a glutamine; at position EU424 a glutamic acid or an arginine, a lysine, or an asparagine; at position EU426 an aspartic acid, a glutamine, an alanine, or a tyrosine; at position EU433 an aspartic acid; at position EU436 a threonine; at position EU438 a glutamic acid, an arginine, a serine, or a lysine; and at position EU440 a glutamic acid, aspartic acid or a glutamine.
  • the FcRn-binding domain comprises: a) at position EU252 a tyrosine, at position EU387 an arginine, at position EU434 a tyrosine, and at position EU436 a valine; or b) at position EU252 a tyrosine, at position EU422 a glutamic acid, at position EU434 a tyrosine, and at position EU436 a valine; or c) at position EU252 a tyrosine, at position EU422 an arginine, at position EU434 a tyrosine, and at position EU436 a valine; or d) at position EU252 a tyrosine, at position EU422 a serine, at position EU434 a tyrosine, and at position EU436 a valine; or e) at position EU252 a
  • a method for improving the pharmacokinetics of an antigen-binding molecule comprising the step of introducing an amino acid substitution into an FcRn-binding domain of said antigen-binding molecules at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • a method for delaying the elimination of an antigen-binding molecule in a subject comprising the step of introducing an amino acid substitution into an FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • a method of prolonging the plasma retention time of an antigen-binding molecule comprising the step of introducing an amino acid substitution into an FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • a method for increasing the plasma antigen-elimination rate of an antigen-binding molecule comprising the step of introducing an amino acid substitution into an FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • a method for increasing the ability of an antigen-binding molecule to eliminate plasma antigen comprising the step of introducing an amino acid substitution into an FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • a method for producing antigen-binding molecules according to any one of [1] to [25], which comprises the steps of (a) selecting a parent FcRn-binding domain and altering the parent FcRn by introducing an amino acid substitution at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; (b) selecting an antigen-binding domain of an antigen-binding molecule and altering at least one amino acid in the antigen-binding domain in order to get a pH-dependent antigen-binding domain or a calcium-ion dependent antigen-binding domain; (c) obtaining a gene encoding an antigen-binding molecule in which the human FcRn-binding domain and the antigen-binding domain prepared in (a) and (b) are linked and (d) producing an antigen-binding molecule using
  • step a) further an amino acid substitution at position EU256 into the FcRn binding domain is introduced.
  • step a) further comprises a step of introducing into the FcRn-binding domain an amino acid substitution at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • An antigen-binding molecule comprising a modified FcRn binding domain, wherein the modified FcRn binding domain comprises an amino acid substitution at one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440, wherein the binding affinity of said antigen-binding molecule for a pre-existing anti-drug antibody (ADA) at a neutral pH is not significantly increased as compared to the binding affinity of antigen-binding molecule comprising an intact FcRn binding domain.
  • ADA anti-drug antibody
  • the antigen-binding molecule according to [38] or [39], wherein the amino acid substituting one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440 is selected from the group consisting of a) at position EU387 an arginine; b) at position EU422 a glutamic acid, an arginine, a serine, aspartic acid, lysine, threonine, or glutamine; c) at position EU424 a glutamic acid, an arginine, a lysine, or asparagines; d) at position EU426 an aspartic acid, a glutamine, an alanine, or a tyrosine; e) at position EU433 an aspartic acid f) at position EU436 a threonine g) at position EU438 a glutamic acid, an arginine, a serine,
  • the modified FcRn binding domain further comprises an amino acid substitution at one or more positions of the FcRn binding domain selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU434, and EU436, wherein said substitutions confer an increase in FcRn binding activity in the neutral pH or acidic pH range.
  • the modified FcRn binding domain comprises three or more amino acid substitutions wherein the three or more substitutions are one of the combinations set forth in Tables 13 and 15.
  • a method for decreasing the binding activity for a pre-existing ADA of an antigen-binding molecule comprising an FcRn binding domain having an increased binding activity for an FcRn at neutral or acidic pH and an increased binding activity for a pre-existing ADA at a neutral pH, said method comprising the steps of a) providing an antigen-binding molecule with an FcRn binding domain having an increased binding activity for FcRn at neutral or acidic pH and an increased binding activity for a pre-existing ADA at a neutral pH; and b) substituting an amino acid in the FcRn binding domain at one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440 to yield
  • step b) comprises substituting an amino acid at three or more positions wherein the three or more positions are one of the combinations set forth in Table 10.
  • step b) comprises introducing three or more the amino acid substitutions into the FcRn-binding domain wherein the three or more the amino acid substitutions are one of the combinations set forth in Table 11.
  • a method for increasing the total number of antigens to which a single antigen-binding molecule can bind without significantly increasing the binding activity for a pre-existing ADA at neutral pH as compared to a parent antibody comprising the steps of a) providing an antigen-binding molecule comprising a parent FcRn binding domain, b) altering the parent FcRn binding domain of step a) by substituting an amino acid in the amino acid sequence of the parent FcRn binding domain at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU42
  • a method for facilitating the extracellular release of an antigen-free antigen-binding molecule taken up into cells in an antigen-bound form without significantly increasing the binding activity of said antigen-binding molecule for a pre-existing ADA at neutral pH as compared to a parent antibody comprising the steps of a) providing an antigen-binding molecule comprising a parent FcRn-binding domain, b) altering the parent FcRn binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, and EU428; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or
  • a method for increasing the ability of an antigen-binding molecule to eliminate plasma antigen without significantly increasing the binding activity for pre-existing ADA at neutral pH compared to parent antibody comprising the steps of a) providing an antigen-binding molecule comprising a parent FcRn-binding domain, b) altering the parent FcRn binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, and EU428; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433,
  • a method for improving the pharmacokinetics of an antigen-binding molecule without significantly increasing the binding activity for a pre-existing ADA at neutral pH as compared to a parent antibody comprising the steps of a) providing an antigen-binding molecule comprising a parent FcRn-binding domain, b) altering the parent FcRn-binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU
  • a method for reducing total or free antigen plasma concentration without significantly increasing the binding activity for a pre-existing ADA at neutral pH as compared to a parent antibody comprising the steps of a) providing an antigen-binding molecule comprising a parent FcRn-binding domain, wherein the antigen-binding molecule comprises an antigen-binding domain which can bind said antigen, b) altering the parent FcRn-binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from
  • a method for producing an antigen-binding molecule comprising an FcRn binding domain having an increased binding activity for an FcRn at neutral or acidic pH and a decreased binding activity for an pre-existing ADA at neutral pH, said method comprising the steps of (a) providing an FcRn binding domain having an increased binding activity for an FcRn at neutral or acidic pH ranges and pre-existing ADA at neutral pH ranges, (b) substituting an amino acid at one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440, (c) selecting an antigen-binding domain of an antigen-binding molecule and altering at least one amino acid in the antigen-binding domain in order to get a pH-dependent antigen-binding domain, or selecting an calcium-ion dependent antigen-binding domain; (d) obtaining a gene encoding an antigen-binding molecule in which the human FcRn-binding domain
  • the FcRn binding domain having an increased binding activity for FcRn and pre-existing ADA at neutral or acidic pH ranges and for pre-existing ADA in the neutral pH ranges comprises an amino acid substitution at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • the amino acid substitutions introduced in step b) are at three or more positions wherein said three or more positions are one of the combinations set forth in Table 10.
  • FIG. 1A shows a schematic depiction of antigen elimination from plasma of an antibody of the prior art (“conventional antibody”) compared with pH-dependent antigen binding antibody with enhanced FcRn, both binding a soluble antigen at neutral pH.
  • the conventional antibody binds to the antigen in the plasma and is non-specifically taken up by cells into acidic endosomes. Under acidic conditions of the endosomes, the conventional antibody binds the FcRn inside a vesicle and is transported back to the surface of the cell where it is again released. The antigen is bound to the antigen-binding domain during the whole internalization and recycling process.
  • a pH-dependent antigen binding antibody with enhanced FcRn binding at neutral pH binds to the FcRn on the surface of the cell and is internalized rapidly into the cell and therefore in a higher frequency than a conventional antibody.
  • the antigen dissociates from the modified antibody and is transferred to the lysosome where it is proteolytically degraded.
  • the antibody, still bound to the FcRn, is recycled back to cell surface. There, the recycled free antibody can bind to another antigen once again.
  • such pH-dependent antigen-binding antibody with improved binding affinity to FcRn at neutral pH can deliver significantly higher amount of antigen to the lysosome than a conventional antibody and therefore can reduce the total antigen concentration in plasma significantly more than a conventional antibody.
  • FIG. 1B shows a schematic representation of the dissociation of a soluble antigen from an IgG antibody with a pH-dependent antigen-binding domain in the endosome. This results in increased antigen elimination, and allows the antibody to bind to another antigen in the plasma.
  • FIG. 2 shows the plot of hFcRn binding affinity (x axis) and Tm of antibodies comprising Fc variants on the y axis (Fc variants F1-F599: open square; Fc variants F600-F1052: closed square).
  • FIG. 3 shows the plot of hFcRn binding affinity (x-axis) and High Molecular Weight (HMW) portion (in %) (y axis) of antibodies comprising Fc variants (Fc variants F1-F599: open square, Fc variants F600-F1050: closed square)
  • FIG. 4 shows the plot of hFcRn binding affinity (x-axis) and immunogenicity score (Epibase score) of antibodies comprising Fc variants (Fc variants F1-F599: open square, Fc variants F600-F1052: closed square).
  • FIG. 5 shows the plot of hFcRn binding affinity (x-axis) and melting Temperature Tm (y axis) of antibodies comprising Fc variants whose hFcRn binding affinity is stronger than 15 nM (Fc variants of F1-F599 with Kd less than or equal to 15 nM: open square, Fc variants of F600-F1052 with Kd less than or equal to 15 nM (Group 1): closed square).
  • FIG. 6 shows the plot of hFcRn binding affinity (x axis) and HMW (in %) (y-axis) of antibodies comprising Fc variants whose hFcRn binding affinity is stronger than 15 nM (Fc variants of F1-F599 with Kd less than or equal to 15 nM: open square; Fc variants of F600-F1052 with Kd less than or equal to 15 nM (Group 1): closed square).
  • FIG. 7 shows the plot of hFcRn binding affinity and immunogenicity score of antibodies comprising Fc variants whose hFcRn binding affinity is stronger than 15 nM (Fc variants of F1-F599 with Kd less than or equal to 15 nM: open square; Fc variants of F600-F1052 with Kd less than or equal to 15 nM (Group 1): closed square).
  • FIG. 17 shows a graphical depiction of the plasma antigen (hsIL-6R) concentration over time in a human FcRn transgenic mouse after injection of Fv-4-IgG1, Fv-4-F652, Fv-4-F890 and Fv-4-F946 and in a control mouse (no antibody injection).
  • hsIL-6R plasma antigen
  • FIG. 18 shows a graphical depiction of the plasma antibody concentration over time in human FcRn transgenic mouse after injection of Fv-4-IgG1, Fv-4-F652, Fv-4-F890 and Fv-4-F946.
  • FIG. 19 shows a graphical depiction of the plasma antigen (hsIL-6R) concentration over time in human FcRn transgenic mouse of control (no antibody injection) and after injection of Fv-4-IgG1, Fv-4-F11 and Fv-4-F652.
  • FIG. 20 shows a graphical depiction of the plasma antibody concentration over time in human FcRn transgenic mouse after injection of Fv-4-IgG1, Fv-4-F11 and Fv-4-F652.
  • FIG. 21 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the humanized anti-IL-6 receptor antibody Fv-4-IgG1 ( FIG. 21-1 ), an YTE variant ( FIG. 21-2 ) and a LS variant ( FIG. 21-3 ) thereof.
  • ECL electrochemiluminescence
  • FIG. 22 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the humanized anti-IL-6 receptor antibody Fv-4-IgG1 ( FIG. 22-1 ), a Fv-4-N434H ( FIG. 22-2 ), Fv-4-F11 ( FIG. 22-3 ), Fv-4-F68 ( FIG. 22-4 ), Fv-4-890 ( FIG. 22-5 ) and Fv-4-F947 ( FIG. 22-6 ).
  • ECL electrochemiluminescence
  • FIG. 23 shows the mean ( FIG. 23-1 ), geomean ( FIG. 23-2 ) and median ( FIG. 23-3 ) of the ECL response of the serum from fifteen individual RA patients against Fv-4-IgG1, Fv-4-F11, Fv-4-F68, Fv-4-F890 and Fv-4-F947 shown in FIG. 22 .
  • FIG. 24 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the humanized anti-IL-6 receptor antibody Fv-4-IgG1 ( FIG. 24-1 ) and of the variants Fv-4-F890, Fv-4-F1058, Fv-4-F1059, Fv-4-F1060, Fv-4-F1061, Fv-4-F1062, Fv-4-F1063, Fv-4-F1064, Fv-4-F1065, Fv-4-F1066, Fv-4-F1067, Fv-4-F1068, Fv-4-F1069, Fv-4-F1070, Fv-4-F1071, Fv-4-F1072, and Fv-4-F1073 ( FIG. 24-2 to FIG. 24-18 ).
  • ECL electrochemiluminescence
  • FIG. 25 shows the mean ( FIG. 25-1 ), geomean ( FIG. 25-2 ) and median ( FIG. 25-3 ) of the ECL response of the serum from fifteen individual RA patients against Fv-4-IgG1, variants Fv-4-F890, Fv-4-F1058, Fv-4-F1059, Fv-4-F1060, Fv-4-F1061, Fv-4-F1062, Fv-4-F1063, Fv-4-F1064, Fv-4-F1065, Fv-4-F1066, Fv-4-F1067, Fv-4-F1068, Fv-4-F1069, Fv-4-F1070, Fv-4-F1071, Fv-4-F1072, and Fv-4-F1073 shown in FIG. 24 .
  • FIG. 26 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the variants Fv-4-F1104 ( FIG. 26-1 ), Fv-4-F1105 ( FIG. 26-2 ), and Fv-4-F1106 ( FIG. 26-3 ).
  • ECL electrochemiluminescence
  • FIG. 27 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the variants Fv-4-F1107, Fv-4-F1108, Fv-4-F1109, Fv-4-F1110, Fv-4-F1111, Fv-4-F1112, Fv-4-F1113, and Fv-4-F1114 ( FIG. 27-1 to FIG. 27-8 )
  • FIG. 28 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the variants Fv-4-F1230 ( FIG. 28-1 ), Fv-4-F1231 ( FIG. 28-2 ), Fv-4-F1232 ( FIG. 28-3 ).
  • ECL electrochemiluminescence
  • FIG. 29 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the variants Fv-4-F947 ( FIG. 29-1 ), Fv-4-F1119 ( FIG. 29-2 ), Fv-4-F1120 ( FIG. 29-3 ), Fv-4-F1121 ( FIG. 29-4 ), Fv-4-F1122 ( FIG. 29-5 ), Fv-4-F1123 ( FIG. 29-6 ), and Fv-4-F1124 ( FIG. 29-7 ).
  • ECL electrochemiluminescence
  • FIG. 30-1 to FIG. 30-4 show the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the variants Fv-4-F939, Fv-4-F1291, Fv-4-F1268, and Fv-4-F1269.
  • FIG. 30-5 to FIG. 30-9 show the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variants Fv-4-F1243, Fv-4-F1245, Fv-4-F1321, Fv-4-F1340, and Fv-4-F1323.
  • FIG. 31 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 15 individual RA patients against the variants Fv-4-F890 ( FIG. 31-1 ) and Fv-4-F1115 ( ⁇ F890+S424N, FIG. 31-2 ).
  • ECL electrochemiluminescence
  • ECL electrochemiluminescence
  • FIG. 33 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variants Fv-4-LS ( FIG. 33-1 ), Fv-4-F1380 ( FIG. 33-2 ), Fv-4-F1384 ( FIG. 33-3 ), Fv-4-F1385 ( FIG. 33-4 ), Fv-4-F1386 (LS+S426Y, FIG. 33-5 ), Fv-4-F1388 ( FIG. 33-6 ), and Fv-4-F1389 (LS+Y436T, FIG. 33-7 ).
  • ECL electrochemiluminescence
  • FIG. 34 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F939.
  • FIG. 35 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1378
  • FIG. 36 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1379
  • FIG. 37 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1262
  • FIG. 38 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1138
  • FIG. 39 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1344
  • FIG. 40 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1349
  • FIG. 41 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1350
  • FIG. 42 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1351
  • FIG. 43 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1261
  • FIG. 44 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1263
  • FIG. 45 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1305
  • FIG. 46 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1306
  • FIG. 47 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1268
  • FIG. 48 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1269
  • FIG. 49 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1413
  • FIG. 50 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1416
  • FIG. 51 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1419
  • FIG. 52 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1420
  • FIG. 53 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1370
  • FIG. 54 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1371
  • FIG. 55 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1599
  • FIG. 56 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1600
  • FIG. 57 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1566
  • FIG. 58 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1448
  • FIG. 59 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1601
  • FIG. 60 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1602
  • FIG. 61 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1603
  • FIG. 62 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1531
  • FIG. 63 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1604
  • FIG. 64 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1605
  • FIG. 65 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1586
  • FIG. 66 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1592
  • FIG. 67 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1610
  • FIG. 68 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1611
  • FIG. 69 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1612
  • FIG. 70 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1613
  • FIG. 71 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1614
  • FIG. 72 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1615
  • FIG. 73 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1567
  • FIG. 74 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1572
  • FIG. 75 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1576
  • FIG. 76 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1578
  • FIG. 77 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1579
  • FIG. 78 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1641
  • FIG. 79 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1642
  • FIG. 80 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1643
  • FIG. 81 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1644
  • FIG. 82 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1645
  • FIG. 83 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1646
  • FIG. 84 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1647
  • FIG. 85 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1648
  • FIG. 86 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1649
  • FIG. 87 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1650
  • FIG. 88 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1651
  • FIG. 89 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1652
  • FIG. 90 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1653
  • FIG. 91 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1654
  • FIG. 92 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1655
  • FIG. 93 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1329
  • FIG. 94 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1331
  • FIG. 95 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1718
  • FIG. 96 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1719
  • FIG. 97 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1720
  • FIG. 98 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1721
  • FIG. 99 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1671
  • FIG. 100 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1670
  • FIG. 101 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1711
  • FIG. 102 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1712
  • FIG. 103 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1713
  • FIG. 104 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1722
  • FIG. 105 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1723
  • FIG. 106 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1724
  • FIG. 107 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1725
  • FIG. 108 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1675
  • FIG. 109 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1714
  • FIG. 110 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1715
  • FIG. 111 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1716
  • FIG. 112 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1717
  • FIG. 113 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1683
  • FIG. 114 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1756
  • FIG. 115 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1757
  • FIG. 116 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1758
  • FIG. 117 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1759
  • FIG. 118 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1681
  • FIG. 119 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1749
  • FIG. 120 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1750
  • FIG. 121 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1751
  • FIG. 122 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1760
  • FIG. 123 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1761
  • FIG. 124 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1762
  • FIG. 125 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1763
  • FIG. 126 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1752
  • FIG. 127 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1753
  • FIG. 128 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1754
  • FIG. 129 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1755
  • FIG. 130 shows the graphical depiction of the electrochemiluminescence (ECL) response of the serum from 30 individual RA patients against the variant F1685
  • FIG. 131 shows a graphical depiction of the plasma antibody concentration over time in human FcRn transgenic mouse after injection of Fv-4-IgG1, Fv-4-F1243, and Fv-4-F1245.
  • FIG. 132 shows a graphical depiction of the plasma antigen (hsIL-6R) concentration over time in a human FcRn transgenic mouse after injection of Fv-4-IgG1, Fv-4-F1243, and Fv-4-F1245 and in a control mouse (no antibody injection).
  • FIG. 133 shows a graphical depiction of the plasma antibody concentration over time in human FcRn transgenic mouse after injection of Fv-4-IgG1, Fv-4-F1389.
  • FIG. 134 shows a sensorgram of SPR analysis. hIgA binding of anti-hIgA antibody was analyzed under different condition (pH, Ca-concentration).
  • FIG. 135 shows a graphical depiction of the plasma antibody concentration over time in human FcRn transgenic mouse after injection of GA2-F760 and GA2-F1331.
  • FIG. 136 shows a graphical depiction of the plasma hIgA concentration over time in human FcRn transgenic mouse after injection of GA2-F760 and GA2-F1331.
  • FIG. 137 shows a graphical depiction of the plasma antibody concentration over time in human FcRn transgenic mouse after injection of 278-F760 and 278-F1331.
  • FIG. 138 shows a graphical depiction of the plasma hIgE(Asp6) concentration over time in human FcRn transgenic mouse after injection of 278-F760 and 278-F1331.
  • antigen-binding molecules e.g. anti-IL6 receptor antibody
  • antigen-binding molecules e.g. anti-IL6 receptor antibody
  • pH-dependent antigen binding bind to antigen within plasma at pH7.4 and dissociate the antigen within acidic endosome at pH6.0
  • ionized calcium concentration-dependent antigen binding bind to antigen within plasma at high ionized calcium concentration and dissociate the antigen within the endosome at low ionized calcium concentration
  • the present invention provides novel amino acid substitutions in the FcRn-binding domain that increase FcRn binding activity of an antigen-binding molecule in the acidic and neutral pH ranges, wherein the FcRn-binding activity in the neutral pH range is stronger than the one of an intact IgG or an antigen-binding molecule comprising an intact FcRn-binding domain (e.g. stronger than 3200 nM).
  • the modified antigen-binding molecules can reduce the total antigen concentration in plasma after its administration more than a control antigen-binding molecule comprising the same antigen-binding domain but an intact human IgG FcRn-binding domain.
  • Fc receptors are proteins on the surface of immune cells such as natural killer cells, macrophages, neutrophils and mast cells. They bind to the Fc (Fragment, crystallizable) region of antibodies that are attached to infected cells or invading pathogens and stimulate phagocytic or cytotoxic cells to destroy microbes, or infected cells by antibody-mediated phagocytosis or antibody-dependent cell-mediated cytotoxicity.
  • FcRn refers to the neonatal Fc receptor that binds IgG, is similar in structure to MHC class I protein and that, in humans, is encoded by the FCGRT gene.
  • FcRn binding domain refers to a protein domain that directly or indirectly binds to the FcRn.
  • the FcRn is a mammalian FcRn, more preferably, a human FcRn.
  • An FcRn binding domain binding directly to an FcRn is an antibody Fc region.
  • regions capable of binding to a polypeptide such as albumin or IgG, which has human FcRn-binding activity can indirectly bind to human FcRn via albumin, IgG, or such.
  • a human FcRn-binding region may be a region that binds to a polypeptide having human FcRn-binding activity.
  • Fc region or “Fc region of an antigen-binding molecule” as used herein refers to an FcRn-binding domain that directly binds to FcRn, preferably to a mammalian FcRn, more preferably to a human FcRn.
  • an Fc region is an Fc region of an antibody.
  • the Fc region is a mammalian Fc region, more preferably a human Fc region.
  • the Fc region of the invention is an Fc region comprising the second and third constant domain of a human immunoglobulin (CH2 and CH3), more preferably the hinge, CH2 and CH3.
  • the immunoglobulin is an IgG.
  • the Fc region is an Fc region of human IgG1.
  • the present invention provides an antigen-binding molecule having a modified FcRn-binding domain wherein said antigen-binding molecule has an increased FcRn-binding activity in a neutral pH range as compared to an antigen-binding molecule having an intact FcRn-binding domain.
  • the present invention provides an antigen-binding molecule having a modified FcRn-binding domain with an amino acid substitution in an FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • the antigen-binding molecule of the present invention may also comprise substitutions at additional positions.
  • the antigen-binding molecule may comprise a substitution at position EU256 in addition to a substitution at the one or more positions mentioned above.
  • the amino acid at position EU256 is substituted with a glutamic acid.
  • binding affinity refers to the strength of non-covalent interaction between two substances as measured by the dissociation constant (KD) of the complex formed by the two substances, unless expressly defined otherwise.
  • a binding protein (or “ligand”) may, for example, have a KD of less than 10 ⁇ 5 , 10 ⁇ 6 , 10 ⁇ 7 or 10 ⁇ 8 M for a particular target molecule, e.g. the FcRn.
  • KD dissociation constant
  • Binding affinity can be determined by a variety of methods including surface plasmon resonance, equilibrium dialysis, equilibrium binding, gel filtration, ELISA, or spectroscopy (e.g., using a fluorescence assay).
  • An increased binding affinity of an FcRn-binding domain for FcRn at a pH range corresponds to a measured increase of the FcRn-binding affinity as compared to the FcRn-binding affinity measured for an intact FcRn-binding domain. Differences in binding affinity of KD (intact)/KD (variant) is at least 1.5, 2, 3, 4, 5, 10, 15, 20, 50, 70, 80, 100, 500, or 1000 fold.
  • An increased binding affinity of an FcRn-binding domain for FcRn can be in the acidic or neutral pH ranges.
  • the term “antigen-binding molecule comprising an intact FcRn binding domain” refers to an antigen-binding molecule comprising an unmodified FcRn-binding domain.
  • the term “intact IgG FcRn-binding domain” as used herein refers to an unmodified FcRn-binding domain of a human IgG.
  • the FcRn-binding domain is the FcRn-binding domain of an intact human IgG.
  • an intact FcRn-binding domain is an intact Fc region.
  • antibody comprising an intact Fc region refers to an antibody comprising an unmodified Fc region.
  • the antibody from which the unmodified Fc region originates is preferably an IgG.
  • an antibody comprising an intact Fc region is an antibody comprising an unmodified Fc region.
  • An antibody comprising an intact Fc region can be an intact human IgG.
  • intact IgG refers to an unmodified IgG and is not limited to a specific class of IgG. This means that human IgG1, IgG2, IgG3, IgG4 or their allotypic variants can be used as “intact human IgG” as long as it can bind to the human FcRn in the acidic pH range.
  • intact IgG is a human IgG1.
  • an intact IgG is an IgG which comprises a wild type Fc region.
  • an increased FcRn-binding activity of antigen-binding molecule in the neutral pH ranges is preferably stronger than KD 3.2 micromolar.
  • the increased FcRn-binding activity in the neutral pH range is stronger than 700 nanomolar, more preferably stronger than 500 nanomolar and most preferably, stronger than 150 nanomolar.
  • An increased FcRn-binding activity of antigen-binding molecule of the present invention in the acidic pH ranges is generally an FcRn-binding activity in the range of about 2-fold to about 100-fold stronger than the FcRn-binding activity of an intact IgG.
  • the increased FcRn-binding activity of antigen-binding molecule in the acidic pH ranges is at least 10-fold stronger than the FcRn-binding activity of an intact IgG. More preferably, the increased FcRn-binding activity of an antigen-binding molecule of the present invention in the acidic pH range is at least 20-fold stronger than the FcRn-binding activity of an intact IgG.
  • neutral pH range typically refer to pH 6.7 to pH 10.0, preferably any pH value within pH 7.0 to pH 8.0, examples of which include pH 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, and 8.0.
  • a particularly preferred acidic pH value is pH 7.4, which approximates plasma (blood) pH in vivo.
  • acidic pH range typically refer to pH 4.0 to pH 6.5, preferably to any pH value within pH 5.5 to pH 6.5, examples of which include pH 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, and 6.5.
  • a particularly preferred acidic pH value ranges from pH 5.8 to pH 6.0, which approximates the pH in early endosome in vivo.
  • EU387 amino acid positions referred to in this application, such as e.g. “EU387” or “position 387”, are, unless otherwise indicated, numbered according to a scheme called the EU numbering system (Kabat, E. A., T. T. Wu, H. M. Perry, K. S. Gottesman, C. Foeler. 1991. Sequences of Proteins of Immunological Interest. No. 91-3242 U.S. Public Health Services, National Institutes of Health, Bethesda) and refer to positions in an FcRn-binding domain, in particular in an Fc region.
  • substitutions are indicated as for example “EU387R” or “EU440E”, wherein the number given after “EU” indicates the position of the substitution according the EU numbering, and the letter after the number is the substituted amino acid given in the one letter code.
  • Substitutions may also be written as (amino acid 1)-position-(amino acid 2) whereby the first amino acid is the substituted amino acid and the second amino acid is the substituting amino acid at the specified position.
  • substitution and substitution of an amino acid refer to a replacement of an amino acid in an amino acid sequence with another one, wherein the latter is different from the replaced amino acid.
  • Methods for replacing an amino acid are well known to the skilled in the art and include, but are not limited to, mutations of the nucleotide sequence encoding the amino acid sequence.
  • a substitution of an amino acid in an FcRn-binding domain refers to a replacement of an amino acid in reference to the amino acid sequence of a parent FcRn-binding domain.
  • a modified FcRn-binding domain already having the desired substitutions is also included in the FcRn-binding domain of the present invention.
  • a parent FcRn-binding domain is an FcRn-binding domain having at the position EU238 an proline, at position EU250 a threonine, at position EU252 a methionine, at position EU254 a serine, at position EU255 an arginine, at position EU256 a threonine, at position EU258 a glutamic acid, at position EU286 an asparagine, at position EU307 a threonine, at position EU308 a valine, at position EU309 a leucine, at position EU311 a glutamine, at position EU315 an asparagine, at position EU387 a proline, at position EU422 a valine, at position EU424 a serine, at position EU426 a serine, at position EU428 a methionine, at position EU433 a histidine, at position EU434 an asparagine, at position EU436 a tyrosine, at position
  • the parent FcRn-binding domain may comprise substitutions at other positions but preferably, the parent FcRn-binding domain is unmodified.
  • the parent FcRn binding domain is an Fc region (parent Fc region).
  • the parent Fc region is derived from a mammalian antibody; more preferably, the parent Fc region is the Fc region of a human antibody.
  • An Fc region of a human antibody is herein referred to as a human Fc region.
  • a parent Fc region is, preferably an intact Fc region, more preferably a human intact Fc region.
  • the parent Fc region is the Fc region of an IgG, more preferably of a human IgG. Even more preferably, a parent Fc region is a human Fc region comprising the wild type hinge, wildtype CH2 and wildtype CH3 domain.
  • the term parent antibody refers to an antibody comprising a parent Fc region.
  • Parent antigen-binding molecules include, but are not limited to, receptor proteins (membrane-bound receptors and soluble receptors), antibodies that recognize a membrane antigen such as cell surface markers, and antibodies that recognize a soluble antigen such as cytokines
  • parent antigen-binding molecule refers to an antigen-binding molecule having a parent FcRn-binding domain.
  • the origin of “parent antigen-binding molecule” is not limited and it may be obtained from any organism: of non-human animals or human.
  • the organism is selected from the group consisting of mouse, rat, guinea pig, hamster, gerbil, cat, rabbit, dog, goat, sheep, cow, horse, camel, and non-human primate.
  • “parent antigen-binding molecule” can also be obtained from cynomolgus monkey, marmoset, rhesus monkey, chimpanzee or human.
  • the parent IgG may be a naturally occurring IgG, or a variant or engineered version of a naturally occurring IgG.
  • Parent IgG may refer to the polypeptide itself, compositions that comprise the parent IgG, or the amino acid sequence that encodes it.
  • parent IgG includes known commercial, recombinantly produced IgG as outlined below.
  • parent IgG is obtained from human IgG1 but not limited to a specific subclass of IgG. This means that human IgG1, IgG2, IgG3, or IgG4 can be appropriately used as “parent IgG”.
  • any subclass of IgGs from any organisms described hereinbefore can be preferably used as “parent IgG”.
  • Example of variant or engineered version of a naturally occurring IgG is described in Curr Opin Biotechnol. 2009 December; 20(6): 685-91, Curr Opin Immunol. 2008 August; 20(4): 460-70, Protein Eng Des Sel. 2010 April; 23(4): 195-202, WO 2009/086320, WO 2008/092117, WO 2007/041635 and WO 2006/105338, but not limited thereto.
  • An FcRn-binding domain or Fc region of the present invention may comprise a substitution at two or more positions which is herein referred to as a “combination” of substitutions.
  • an Fc region defined by the combination “EU424/EU434/EU436” is an Fc region that comprises a substitution at the positions EU424, EU434 and EU436.
  • the substituting amino acid may be any amino acid unless specifically mentioned herein, including but not limited to the group consisting of: alanine (Ala, A), arginine (arg, R), asparagines (asn, N), aspartic acid (asp, D), cysteine, (cys, C), glutamic acid (glu, E), glutamine (gln, Q), glycine (gly, G), histidine (his, H), isoleucine (ile, I), leucine (leu, L), lysine (lys, K), methionine (met, M), phenylalanine (phe, F), proline (pro, P), serine (ser, S), threonine (thr, T), tryptophan (trp, W), tyrosine (tyr, Y), and valine (val, V).
  • alanine Al, A
  • arginine arg, R
  • asparagines asn, N
  • the substituting amino acid at any one of the positions EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440 is selected from the group consisting of: alanine (Ala, A), arginine (arg, R), glutamic acid (glu, E), glutamine (gln, Q), aspartic acid (asp, D), serine (ser, S), threonine (thr, T), tyrosine (tyr, Y), and lysine (lys, K).
  • the antigen-binding molecule of the present invention has a modified FcRn-binding domain comprising an amino acid substitution with an amino acid different from the substituted one
  • the substituting amino acid may be any amino acid unless specifically mentioned herein.
  • Preferred substituting amino acids for the positions EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 are shown in Table 1.
  • the modified FcRn-binding domain of the present invention comprises at least one of amino acids substitutions set forth in Table 1. It is possible to use the FcRn-binding domains without any alteration as long as they already have at least one of the above given amino acids at the specified position and said FcRn-binding domain has human FcRn-binding activity in the acidic and neutral pH ranges, whereby the FcRn-binding activity in the neutral pH ranges is increased.
  • the modified antigen-binding molecule of the present invention comprises a modification at three or more positions in the FcRn-binding domain, wherein the three or more positions are one of the combinations set forth in Tables 2, 4 to 7.
  • the antigen-binding molecule of the present invention comprises three or more amino acid substitutions in a FcRn-binding domain, wherein the three or more substitutions are one of the combinations set forth in Tables 3, 12, 14, and 17 to 20.
  • immunogenicity of a protein pharmaceutical in human is important, since the presence of anti-drug antibodies would result in clearance of the drug from the body and thus loss of therapeutic efficacy (IDrugs 2009; 12:233-7.).
  • substitutions are introduced into a wild type Fc domain (such as IgG1 Fc domain), the modified sequence becomes a non-human sequence.
  • a modified sequence could be presented by MHC class II and therefore could be immunogenic in human patients.
  • Proteins will not be developed as a drug if they comprise Fc variants that exhibit poor stability and purity, and poor immunogenicity would hinder clinical development. It is therefore an objective of the present invention to improve the FcRn binding affinity at pH7.4 without
  • the present invention also provides an antigen-binding molecule comprising an amino acid substitution in the FcRn-binding domain at the positions EU252, EU434, EU307 and EU311, having a binding activity for the FcRn at pH 7 of more than 15 nM, a melting temperature Tm of 57.5 degrees C. or higher, an HMW of less than 2% and a low immunogenicity wherein a low immunogenicity is equivalent to a score of less than 500 determined with Epibase (Lonza).
  • an antigen-binding molecule comprising an amino acid substitution in the FcRn-binding domain at the positions EU252, EU434, EU307 and EU311, having a binding activity for the FcRn at pH 7 of more than 15 nM, a melting temperature Tm of 57.5 degrees C. or higher, an HMW of less than 2% and a low immunogenicity wherein a low immunogenicity is equivalent to a score of less than 500 determined with Epibase (Lonza).
  • an antigen-binding molecule comprising an amino acid substitution in the FcRn-binding domain at four or more positions, wherein the four or more positions are one of the combinations of the group consisting of
  • EU252/EU434/EU307/EU311/EU436 in combination with one or more positions selected from the group consisting of EU286, EU308, and EU428.
  • the modified FcRn-binding domain comprises:
  • the present invention also provides an antigen-binding molecule comprising an amino acid substitution in the FcRn-binding domain at three or more positions, wherein said three or more positions are one of the combinations of the group consisting of a) EU252/EU434/EU307/EU311; and b) EU252/EU434/EU308; wherein the FcRn-binding activity of said antigen-binding molecule at neutral pH is 15 to 50 nM, the Tm is higher than 60 degrees C., an HMW of less than 2% and wherein the antigen-binding molecule has a low immunogenicity whereby a low immunogenicity is equivalent to a score of less than 500 determined with Epibase (Lonza).
  • amino acid substitutions are at four or more positions wherein the four or more positions are one of the combinations set forth in Table 5.
  • EU252/EU434/EU307/EU311/EU286 b) EU252/EU434/EU307/EU311/EU286/EU254 c) EU252/EU434/EU307/EU311/EU436 d) EU252/EU434/EU307/EU311/EU436/EU254 e) EU252/EU434/EU307/EU311/EU436/EU250 f) EU252/EU434/EU308/EU250 g) EU252/EU434/EU308/EU250/EU436/ h) EU252/EU434/EU308/EU250/EU307/EU311
  • an antigen-binding molecule comprising four or more amino acid substitutions wherein the four or more substitutions are one of the combinations of the group consisting of:
  • the present invention also provides an antigen-binding molecule comprising an amino acid substitution in the FcRn-binding domain
  • amino acid substitutions are at three or more positions, wherein the three or more positions are one of the combinations set forth in Table 6.
  • the modified antigen-binding molecule comprises three or more amino acid substitutions wherein the three or more substitutions are one of the combinations of the group consisting of:
  • the present invention further provides an antigen-binding molecule that comprises an amino acid substitution in the FcRn-binding domain at three or more positions, wherein the three or more positions are one of the combinations set forth in Table 7.
  • Said modified antigen-binding molecules have a binding activity for the FcRn at pH 7 of 150 to 700 nM, a Tm of higher than 66.5 degrees C., an HMW of less than 2% and a very low immunogenicity, wherein a very low immunogenicity is defined as a score of less than 250 determined with Epibase (Lonza).
  • the modified antigen-binding molecules comprise three or more substitutions wherein the three or more substations are one of the combinations of the group consisting of
  • Substitutions of amino acids in an antibody can yield negative consequences, for example an increase in the immunogenicity of the therapeutic antibody which, in turn, can result in a cytokine storm and/or production of anti-drug antibodies (ADAs). Since ADAs can influence the efficacy and pharmacokinetics of therapeutic antibodies and sometimes lead to serious side effects, the clinical utility and efficacy of the therapeutic antibodies can be limited. Many factors influence the immunogenicity of therapeutic antibodies, and the presence of effector T-cell epitopes is one of the factors. Likewise, the presence of pre-existing antibodies against a therapeutic antibody can also be problematic.
  • rheumatoid factor an auto-antibody (an antibody directed against a self protein) against the Fc portion of an antibody (i.e. IgG).
  • the rheumatoid factor is found in particular in patients suffering of systemic lupus erythematosus (SLE) or rheumatoid arthritis. In arthritis patients, RF and IgG join to form immune complexes that contribute to the disease process. Recently, it was reported that a humanized anti-CD4 IgG1 antibody having an Asn434H is mutation elicited significant rheumatoid factor binding (Clin Pharmacol Ther.
  • RF is a polyclonal auto-antibody against human IgG, and the epitope of the RF in the sequence of the human IgG varies among the clones, but the RF epitope seems to be located in the CH2/CH3 interface region as well as CH3 domain which could overlap with the FcRn binding epitope. Therefore, mutations to increase the binding affinity to FcRn at neutral pH might also increase the binding affinity to specific clone of RF.
  • present invention also provides antigen-binding molecules comprising a modified FcRn-binding domain (preferably a modified Fc region), whereby the binding activity for a pre-existing ADA at a neutral pH is not significantly increased as compared to the binding affinity of an antigen-binding molecule comprising a wild type Fc region.
  • the modified FcRn-binding domain preferably comprises an amino acid substitution at one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • substitutions are preferably introduced in an FcRn-binding domain or Fc region of an antigen-binding molecule that has increased affinity for the FcRn at neutral or acidic pH whereby said modified FcRn-binding domain or Fc region has an increased binding activity for a pre-existing ADA at neutral pH.
  • the effect of the substitutions is a decrease of the binding activity for the pre-existing ADA.
  • a modified FcRn-binding domain or a modified Fc region of the present invention has a decreased binding activity to a pre-existing ADA as compared to an FcRn binding domain or an Fc region that has an increased binding activity to the FcRn at neutral or acidic pH, and an increased binding activity to the pre-existing anti-drug antibody in the neutral pH ranges.
  • an antigen-binding molecule having an increased binding activity at neutral pH for the FcRn and a pre-existing ADA are the antigen-binding molecules comprising an amino acid substitution at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 as described above. It may also comprise a substitution at position EU256 in addition to a substitution at the one or more positions mentioned above.
  • the amino acid at position EU256 is substituted with a glutamic acid.
  • the present invention also provides antigen-binding molecules comprising a modified Fc region which has an increased affinity for FcRn at neutral or acidic pH whereby the affinity for a pre-existing anti-drug antibody (ADA) at a neutral pH is not significantly increased compared to the binding affinity of antigen-binding molecule comprising a wild type Fc region.
  • the present invention provides an antigen-binding molecule comprising a modified Fc region with an increased affinity for FcRn at neutral or acidic pH which comprises an amino acid substitution at one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the antigen-binding molecule comprising a modified Fc region with an increased affinity for FcRn at neutral or acidic pH, whereby the binding activity at neutral pH for a pre-existing ADA is not significantly increased as compared to a control antigen-binding molecule, wherein the modified Fc region comprises an amino acid substitution at one or more of the positions selected from the substitutions as shown in Table 8.
  • anti-drug antibody and “ADA” as used herein refers to an endogenous antibody that has binding affinity for an epitope located on a therapeutic antibody and is thus capable of binding said therapeutic antibody.
  • pre-existing anti-drug antibody and “pre-existing ADA” as used herein refers to an anti-drug antibody that is present and detectable in the blood of a patient prior to the administration of the therapeutic antibody to the patient.
  • the pre-existing ADA is a human antibody.
  • the pre-existing ADA is the rheumatoid factor, a polyclonal or monoclonal autoantibody against the Fc region of human IgG antibody.
  • the epitopes of rheumatoid factor are located in the CH2/CH3 interface region as well as the CH3 domain but can vary among clones.
  • An antigen-binding molecule comprising an FcRn-binding domain region (or an Fc region) that has an increased affinity for FcRn at neutral or acidic pH and for a pre-existing anti-drug antibody at neutral pH is an antigen-binding molecule comprising an FcRn-binding domain (or an Fc region) that was modified to increase the binding affinity of the FcRn-binding domain (or Fc region) of an antigen-binding molecule for FcRn as compared to an antibody comprising an intact FcRn-binding domain (or intact Fc region).
  • Modifications contemplated include, but are not limited to, substitutions of the amino acids in the amino acid sequence of the Fc portion of an antigen-binding domain.
  • the antigen-binding molecule comprising an FcRn-binding domain or an Fc region, which has an increased binding activity for a) a pre-existing ADA in a neutral pH range and for FcRn at neutral (in case of an antigen-binding molecule of interest having an increased FcRn-binding activity at a neutral pH) or acidic pH (in case of an antigen-binding molecule of interest having an increased FcRn-binding activity at an acidic pH) is referred herein as “Reference Antibody”.
  • a “Reference Antibody” is preferably the modified antigen-binding molecule before substituting an amino acid at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440, more preferably before introducing any one of the substitutions set for in Table 8.
  • a “Reference Antibody” may be an antigen-binding molecule comprising an amino acid substitution in an FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • An example for a “Reference Antibody” having an increased FcRn-binding activity in the neutral pH ranges is an antigen-binding molecule comprising an Fc region with increased affinity for FcRn in the neutral pH ranges and having increased affinity for a pre-existing ADA at neutral pH comprising an amino acid substitution in the Fc region at
  • the antigen-binding molecule comprising an Fc region with increased affinity for FcRn in the neutral pH ranges and having increased affinity for a pre-existing ADA at neutral pH ranges comprises one of the combinations set forth in Table 9.
  • An example for a “Reference Antibody” having an increased FcRn-binding activity in the acidic pH ranges is an antigen-binding molecule comprising an Fc region with increased affinity for FcRn in the acidic pH ranges and having increased affinity for a pre-existing ADA at neutral pH ranges preferably comprise a substitution
  • the antigen-binding molecule comprising an Fc region with increased affinity at acidic pH ranges and having increased affinity for a pre-existing ADA at neutral pH comprises
  • the antigen-binding molecule comprising an Fc region which comprises one of the following substitutions or combinations a) M252Y/S254T/T256E, b) M428L/N434S or c) T250Q and M428L or d) M434H (EU numbering) has an increased binding activity to the FcRn at acidic pH without increasing the binding activity in the neutral pH ranges.
  • the binding activity of an Fc region of antigen-binding molecule for a pre-existing anti-drug antibody is expressed in the present application as an electrochemiluminescence (ECL) response at neutral pH; however, there are other suitable methods for determining the binding activity for a pre-existing ADA known to the skilled in the art.
  • ECL assay is for example described in Moxness et al (Clin Chem, 2005, 51:1983-85) and in the EXAMPLES of the present invention. Conditions used in the assay for determining the binding activity for a pre-existing ADA can be appropriately selected by those skilled in the art, and thus are not particularly limited.
  • An increased or higher binding affinity for a pre-existing ADA is increased as compared to the binding affinity for the pre-existing ADA of a Control Antigen-binding Molecule.
  • Control Antigen-binding Molecule refers to an antigen-binding molecule comprising an intact human Fc region, preferably an antibody or antibody derivative comprising an intact human Fc region.
  • the binding affinity for a pre-existing ADA may be assessed at any temperature from 10 degrees Celsius to 50 degrees Celsius.
  • a temperature at from 15 degrees Celsius to 40 degrees Celsius is employed in order to determine the binding affinity between human Fc region and human pre-existing ADA.
  • any temperature at from 20 degrees Celsius to 35 degrees Celsius is employed in order to determine the binding affinity between human Fc region and human pre-existing ADA.
  • the temperature is between 20 and 25 degrees C., more preferably at 25 degrees C.
  • the interaction between human pre-existing ADA and human Fc region is measured at pH 7.4 (or pH7.0) and at 25 degrees C.
  • an increased binding affinity for a pre-existing ADA refers to a measured increase in binding affinity (i.e., KD) of an antigen-binding molecule of the present invention for a pre-existing ADA as compared to the binding affinity measured of a Control Antigen-binding Molecule for the pre-existing ADA.
  • KD binding affinity
  • Such an increase in binding affinity for a pre-existing ADA can be observed in an individual patient or in a patient group.
  • a patient is a person suffering from an autoimmune disease. More preferably, a patient is a person suffering from an arthritic disease or systemic lupus erythematosus (SLE). Arthritic diseases include in particular rheumatoid arthritis.
  • a significant increase of the binding activity for a pre-existing ADA in an individual patient corresponds to a measured increase of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60% of the binding activity for a pre-existing ADA of a therapeutic antigen-binding molecule (i.e. a therapeutic antibody) comprising a modified Fc region in a patient as compared to the binding affinity for the pre-existing ADA of a Control Antigen-binding Molecule.
  • a therapeutic antigen-binding molecule i.e. a therapeutic antibody
  • the increase is at least 20%, more preferably the increase is at least 30%, even more preferably, it is at least 40% and most preferably the increase is at least 50% of the binding activity of a antigen-binding molecule comprising a modified Fc region as compared to the binding affinity for the pre-existing ADA of a control antigen-binding molecule.
  • a significant increase in the binding activity of an antigen-binding molecule for a pre-existing ADA in a patient is preferably an ECL response to the antigen-binding molecule of more than 250, preferably to an ECL of at least 500, more preferably to an ECL of at least 1000, most preferably to an ECL of at least 2000.
  • the increase is an increase as compared with the ECL response of a Control Antigen-binding Molecule of less than 500 (preferably of less than 250).
  • Preferred ranges between an the binding activity for a pre-existing ADA of the Control Antigen-binding Molecule and that of an antigen-binding molecule with a modified Fc region are in particular ECL responses from less than 250 to at least 250, from less than 250 to at least 500, from less than 500 to 500 or more, from less than 500 to 1000 or more, and from less than 500 to at least 2000.
  • the increase in the binding activity for a pre-existing ADA may also correspond to a measured increase in the portion of patients in a patient population having an ECL response of at least 500 (preferably at least 250) to an the antigen-binding molecule with an increased binding activity to a) the FcRn at neutral or acidic pH and b) an pre-existing ADA at neutral pH as compared to the portion of patients having an ECL response of at least 500 (preferably at least 250) at neutral pH to a control antigen-binding molecule.
  • a “significant” increase in the portion of patients in a patient population is preferably an increase of at least 10%, at least 20%, at least 30%, at least 40%, at least 50% patients having a ECL response of the therapeutic antigen-binding molecule comprising a modified Fc region to the rheumatoid factor at neutral pH of 500 or less (preferably of 250 or more) compared to the portion of patients having an ECL response to a Control Antigen-binding Molecule.
  • the increase is at least 20%, more preferably at least 30%, even more preferably, it is at least 40% and most preferably it is 50% or more.
  • a decrease in the binding affinity for a pre-existing ADA refers to a measured decrease in binding activity (i.e., KD or ECL response) as compared to the binding activity measured for a Reference Antibody,
  • binding affinity for a pre-existing ADA can be observed in an individual patient or in a patient group.
  • the decrease of the affinity of a therapeutic antigen-binding molecule for a pre-existing ADA at neutral pH in an individual patient refers to a measured decrease at neutral pH in the binding activity as compared to the binding activity measured for a Reference Antibody for the pre-existing ADA at neutral pH in said patient.
  • a significant decrease in an individual patient is a measured decrease at neutral pH of at least 10%, at least 20%, at least 30%, at least 40%, at least 50% in the binding activity of the modified antigen-binding molecule for a pre-existing ADA as compared to the binding activity of a Reference Antibody for a pre-existing ADA at neutral pH. More preferably, the decrease is at least 30%, even more preferably, it is 40% and most preferably it is 50% or more as compared to a Reference antibody.
  • the significant decrease in an individual patient of a modified antigen-binding molecule's binding activity for a pre-existing ADA may be measured as a decrease of the ECL response of said antigen-binding molecule as compared with the ECL response of a Reference Antibody from an ECL response of 500 or more, (preferably, from an ECL of 1000 or more, most preferably from an ECL of 2000 or more), to less than 500, preferably of less than 250.
  • Preferred decreases are from an ECL response of 500 or more to an ECL response of less than 500, more preferably from at least 250 to less than 250, even more preferably from at least 500 to less than 250.
  • Preferred ranges are, in particular, from at least 250 to less than 250, from at least 500 to less than 250, from at least 1000 to less than 250, from at least 2000 to less than 250, from at least 500 to less than 500, from at least 1000 to less than 500, and from at least 2000 to less than 500.
  • the decrease may also be a decrease in the percentage of patients in a patient population that has an increased binding of their pre-existing ADA to the modified antigen-binding molecule in neutral pH ranges.
  • the decrease may be measured as a decrease of the percentage of people having an ECL response of their pre-existing ADA to a modified antigen-binding molecule as compared to the ECL response to a Reference Antibody.
  • a decrease may be a decrease of at least 10%, at least 20%, at least 30%, at least 40%, at least 50% in the portion of patients in a patient population in which the therapeutic antigen-binding molecule has an increased binding activity to a pre-existing ADA as compared to the portion of patients having an increased binding activity of the Reference antibody to the pre-existing ADA, wherein the increased binding is expressed as an ECL response of 500 or more, preferably 250 or more.
  • the decrease is at least 20%, more preferably the decrease is at least 30%, even more preferably, it is 40% and most preferably it is 50% or more.
  • a therapeutic antigen-binding molecule of the present invention has low binding activity for a pre-existing ADA at a neutral pH.
  • the binding activity of a modified antigen-binding molecule of the present invention for a pre-existing ADA at a neutral pH is preferably significantly decreased compared to the binding activity of a Reference Antibody for a pre-existing ADA at neutral pH.
  • the binding activity of a modified antigen-binding molecule of the present invention for a pre-existing ADA at a neutral pH is not significantly increased as compared to the binding affinity of a Control Antigen-binding Molecule (has about the same binding activity for a pre-existing ADA as Control Antigen-binding Molecule).
  • a low binding activity or baseline affinity for a pre-existing ADA is preferably an ECL response of less than 500 in a individual patient.
  • a ECL response is less than 250.
  • a low binding activity for a pre-existing ADA is an ECL response of less than 500 in 90% of the patients in the patient population, more preferably in 95% of the patients, most preferably in 98% of the patients.
  • the antigen-binding molecule comprising a modified FcRn-binding domain with an increased affinity for FcRn at neutral or acidic pH, wherein the binding activity at neutral pH for a pre-existing ADA is not significantly increased as compared to a control antigen-binding molecule, whereby the modified FcRn-binding domain of the present invention comprises a substitution at one or more of the positions or combinations set forth in Table 10.
  • the antigen-binding molecule of the present invention comprises a modified FcRn-binding domain having one or more of the substitutions or combinations set forth in Table 11.
  • the antigen-binding molecule comprising a modified FcRn-binding domain with a) an increased affinity for FcRn at neutral or acidic pH b) a binding affinity for a pre-existing ADA at neutral pH which is not significantly increased compared to a Control Antigen-binding Molecule, said antigen-binding molecule comprises any one of the combinations of substitutions set forth in Table 12.
  • an antigen-binding molecule having an increased FcRn binding activity at neutral pH ranges and a binding affinity for a pre-existing ADA at neutral pH that is not significantly increased as compared to an antigen-binding molecule comprising a wild type Fc region comprises an amino acid substitution in an FcRn-binding domain at
  • the substitutions are selected from among the substitutions set forth in Table 11.
  • preferred modified antigen-binding molecules having an increased FcRn-binding activity in neutral pH ranges whereby the binding affinity at neutral pH for a pre-existing ADA is not significantly increased comprises three or more substitutions in the FcRn-binding domain, wherein the three or more substitutions are any one of the combinations no. (2) to (26) and (28) to (59) set forth in Table 12.
  • the present invention also provides an antigen-binding molecule having an increased binding activity for the FcRn at acidic pH ranges and a binding affinity for a pre-existing ADA at neutral pH that is not significantly increased as compared to a Control Antigen-binding Molecule, comprising an amino acid substitution at a) position EU424 or b) position EU438/EU440.
  • substitutions are selected among a) EU424N and EU438R/EU440E.
  • an FcRn-binding domain of an antigen-binding molecule that has an increased binding activity for the FcRn at acidic pH ranges and a binding affinity for a pre-existing ADA at neutral pH that is not significantly increased as compared to a Control Antigen-binding Molecule, comprises one of the substitution combinations set forth in Table 13. More preferably, the antigen-binding molecule having an increased FcRn-binding activity in the acidic pH ranges whereby the binding affinity for a pre-existing ADA at neutral pH that is not significantly increased as compared to a Control Antigen-binding Molecule, comprises any one of the substitution combinations no. (13) to (28) set forth in Table 13.
  • the Fc region of the present invention may also comprise further substitution of an amino acid at one or more of the following positions: EU248, EU249, EU250, EU251, EU252, EU253, EU254, EU255, EU256, EU257, EU305, EU306, EU307, EU308, EU309, EU310, EU311, EU312, EU313, EU314, EU342, EU343, EU344, EU345, EU346, EU347, EU348, EU349, EU350, EU351, EU352, EU380, EU381, EU382, EU383, EU384, EU385, EU386, EU388, EU414, EU415, EU416, EU417, EU418, EU419, EU420, EU421, EU423, EU425, EU427, EU428, EU429, EU430, EU431, EU432, EU433, EU434, EU435, EU436, EU437
  • Substituting an Fc region at any one of these positions may reduce the binding affinity for a pre-existing ADA, in particular for the rheumatoid factor, without negatively affecting the binding affinity for FcRn.
  • the methods of the present invention may further comprise the step of substituting the Fc region of the antigen-binding molecule as described above at one or more of the following positions:
  • a modified antigen-binding molecule that does not bind effector receptors such as Fc gamma RIIa receptor is safer and/or more effective. Therefore, in a preferred embodiment, the modified antigen-binding molecules of the present invention additionally have a weak binding activity for an effector receptor or do not bind to an effector receptor. Examples of an effector receptor include but are not limited to activating Fc gamma receptors, in particular Fc gamma receptor I, Fc gamma receptor II and Fc gamma receptor III.
  • Fc gamma receptor I includes Fc gamma receptor Ia, Fc gamma receptor Ib, and Fc gamma receptor Ic, and subtypes thereof.
  • Fc gamma receptor II includes Fc gamma receptor IIa (which has two allotypes R131 and H131) and Fc gamma receptor IIb.
  • Fc gamma receptor III includes Fc gamma receptor IIIa (which has two allotypes: V158 and F158) and Fc gamma receptor IIIb (which has two allotypes: Fc gamma IIIb-NA1 and Fc gamma IIIb-NA2).
  • Antibodies that have a weak binding activity for effector receptors or do not bind to them are for examples antibodies comprising a silent Fc region or antibodies without an Fc region (e.g. Fab, F(ab)′2, scFv, sc(Fv)2, diabodies).
  • Fc regions having a weak or no binding activity for effector receptors are e.g. described in Strohl et al. (Current Opinion in Biotechnology (2009) 20(6), 685-691). In particular it describes for example deglycosylated Fc regions (N297A, N297Q), and examples of a silent Fc region, which are Fc regions engineered for silenced (or immunosuppressive) effector functionality (IgG1-L234A/L235A, IgG1-H268Q/A330S/P331S, IgG1-C226S/C229S, IgG1-C226S/C229S/E233P/L234V/L235A, IgG1-L234F/L235E/P331S, IgG2-V234A/G237A, IgG2-H268Q/V309L/A330S/A331S, IgG4-L235A/G237
  • WO2008/092117 discloses antibodies comprising silent Fc regions that comprise the substitutions G236R/L328R, L235G/G236R, N325A/L328R, or N325L/L328R (positions according to the EU numbering system). Furthermore, WO 2000/042072 discloses antibodies comprising silent Fc regions which comprise substitutions at one or more of the positions EU233, EU234, EU235, and EU237. WO 2009/011941 discloses antibodies comprising silent Fc regions which comprise deletion of residues from EU231 to EU238.
  • weak binding for effector receptors refers to a binding activity that is 95% or less, preferably 90% or less, 85% or less, 80% or less, 75% or less, more preferably 70% or less, 65% or less, 60% or less, 55% or less, 50% or less, 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 9% or less, 8% or less, 7% or less, 6% or less, 5% or less, 4% or less, 3% or less, 2% or less, 1% or less of the binding activity of an intact IgG (or an antibody comprising an intact Fc region) for the effector receptor.
  • the binding activity to an Fc gamma R preferably reduced by a factor of at least about 10 fold or more, about 50-fold or more, about 100-fold or more as compared with the binding activity of an intact IgG (or an antibody comprising an intact Fc region) for the effector receptor.
  • a silent Fc region is a modified Fc region comprising one or more amino acid substitutions, insertions, additions and/or deletions which reduce the binding for an effector receptor as compared to an intact Fc region.
  • the binding activity for an effector receptor may be so much reduced that the Fc region does not bind an effector receptor anymore.
  • Examples of a silent Fc region include but are not limited to Fc regions which comprise an amino acid substitution at one or more of the positions selected from the group consisting of: EU234, EU235, EU236, EU237, EU238, EU239, EU265, EU266, EU267, EU269, EU270, EU271, EU295, EU296, EU297, EU298, EU300, EU324, EU325, EU327, EU328, EU329, EU331, and EU332.
  • a silent Fc region has a substitution at one or more the positions selected from the group consisting of EU234, EU235, EU236, EU237, EU238, EU239, EU265, EU266, EU267, EU269, EU270, EU271, EU295, EU296, EU297, EU298, EU300, EU324, EU325, EU327, EU328, EU329, EU331, and EU332 with an amino acid selected from the list below.
  • a silent Fc region has a substitution at one or more positions selected from the group consisting of EU235, EU237, EU238, EU239, EU270, EU298, EU325, and EU329 with an amino acid selected from the list below.
  • the amino acid at position EU234 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Arg, Asn, Asp, Gln, Glu, Gly, His, Lys, Met, Phe, Pro, Ser, and Thr.
  • the amino acid at position EU235 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Asn, Asp, Gln, Glu, Gly, His, Ile, Lys, Met, Pro, Ser, Thr, Val and Arg.
  • the amino acid at position EU236 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Asn, Gln, His, Leu, Lys, Met, Phe, Pro and Tyr.
  • the amino acid at position EU237 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Asn, Asp, Gln, Glu, His, Ile, Leu, Lys, Met, Pro, Ser, Thr, Val, Tyr and Arg.
  • the amino acid at position EU238 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Asn, Gln, Glu, Gly, His, Ile, Lys, Thr, Trp and Arg.
  • the amino acid at position EU239 is preferably replaced with one of an amino acid selected from the group consisting of: Gln, His, Lys, Phe, Pro, Trp, Tyr and Arg.
  • the amino acid at position EU265 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Arg, Asn, Gln, Gly, His, Ile, Leu, Lys, Met, Phe, Ser, Thr, Trp, Tyr and Val.
  • the amino acid at position EU266 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Arg, Asn, Asp, Gln, Glu, Gly, His, Lys, Phe, Pro, Ser, Thr, Trp and Tyr.
  • the amino acid at position EU267 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, His, Lys, Phe, Pro, Trp and Tyr.
  • the amino acid at position EU269 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Arg, Asn, Gln, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr and Val.
  • the amino acid at position EU270 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Arg, Asn, Gln, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr and Val.
  • the amino acid at position EU271 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, His, Phe, Ser, Thr, Trp and Tyr.
  • the amino acid at position EU295 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Asn, Asp, Gly, His, Phe, Ser, Trp and Tyr.
  • the amino acid at position EU296 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Gly, Lys and Pro.
  • amino acid at position EU297 is preferably replaced with Ala
  • the amino acid at position EU298 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Gly, Lys, Pro, Trp and Tyr.
  • the amino acid at position EU300 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Lys and Pro.
  • the amino acid at position EU324 is preferably replaced with Lys or Pro.
  • the amino acid at position EU325 is preferably replaced with one of an amino acid selected from the group consisting of: Ala, Arg, Gly, His, Ile, Lys, Phe, Pro, Thr, Trp, Tyr, and Val.
  • the amino acid at position EU327 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Gln, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr and Val.
  • the amino acid at position EU328 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Asn, Gly, His, Lys and Pro.
  • the amino acid at position EU329 is preferably replaced with one of an amino acid selected from the group consisting of: Asn, Asp, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Ser, Thr, Trp, Tyr, Val and Arg.
  • the amino acid at position EU330 is preferably replaced with Pro or Ser.
  • the amino acid at position EU331 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Gly and Lys.
  • the amino acid at position EU332 is preferably replaced with one of an amino acid selected from the group consisting of: Arg, Lys and Pro.
  • a silent Fc region comprises a substitution at position EU235 with Lys or Arg, EU237 with Lys or Arg, EU238 with Lys or Arg, EU239 with Lys or Arg, EU270 with Phe, EU298 with Gly, EU325 with Gly or EU329 with Lys or Arg. More preferably, a silent Fc region comprises a substitution at position EU235 with arginine and at position EU239 with lysine. More preferably, it comprises the substitutions L235R/S239K.
  • the modified antigen-binding molecules of the present invention are preferably deglycosylated. More preferably, the modified antigen-binding molecule of the present invention comprises a mutation at a heavy chain glycosylation site to prevent glycosylation at the site such as e.g. described in WO2005/03175.
  • the modified aglycosyl antigen-binding molecules are prepared by modifying the heavy chain glycosylation site, i.e., introducing the substitution N297Q or N297A (position according to EU numbering system), and expressing the protein in an appropriate host cell. For introducing a substitution a method as described in the EXAMPLES can be used.
  • the modified antigen-binding molecules of the present invention thereof have a weak binding activity for a complement protein or do not bind to a complement protein.
  • the complement protein is C1q.
  • a weak binding activity for a complement protein is preferably a binding activity for a complement protein which is reduced by a factor of about 10 fold or more, about 50-fold or more, about 100-fold or more as compared to the binding activity for a complement protein of an intact IgG or an antibody comprising an intact Fc region.
  • the binding activity of an Fc region for a complement protein can be reduced by modification of the amino acid sequence such as amino acid substitutions, insertions, additions and/or deletions
  • the antigen-binding molecule has an increased FcRn-binding affinity in the acidic or neutral pH and has a weak or no binding activity for an effector receptor and/or a complement protein.
  • such an antigen-binding molecule comprises a substitution in the FcRn-binding domain at
  • the modified antigen-binding molecule of the present invention having a reduced or no binding activity for effector receptors and/or complement proteins comprises one or more substitutions in the Fc regions selected from the group consisting of a substitution at position EU235 with Lys or Arg, at position EU237 with Lys or Arg, at position EU238 with Lys or Arg, at position EU239 with Lys or Arg, at position EU270 with Phe, EU298 with Gly, at position EU325 with Gly and at position EU329 with Lys or Arg. Even more preferably, it comprises a substitution in the Fc region at position EU235 with Arg and at position EU239 with Lys. And even more preferably, it comprises the substitution combination L235R/S239K in the Fc region.
  • the antigen-binding molecule of the present invention having a reduced or no binding activity for effector receptor(s) and/or complement proteins further comprises an amino acid substitutions at c) one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the modified antigen-binding molecules comprise three or more amino acid substitutions in the FcRn-binding domain, wherein the three or more substitutions are one of the combinations set forth in Tables 14 and 15.
  • the antigen-binding molecule of the present invention comprises in addition to the modifications described above, at the position EU257 of the FcRn-binding domain not an amino acid selected from the group consisting of: alanine, valine, isoleucine, leucine, and threonine,
  • the preferred antigen-binding molecule of the present invention comprises in addition to any of the modifications described above at positions EU257 an alanine, a valine, an isoleucine, aleucine, a threonine, an arginine, an asparagine, an aspartic acid, a cysteine, a glutamic acid, a glutamine, a glycine, a histidine, a lysine, a methione, a phenylalanine, a proline, a serine, a tryptophan, or a tyrosine, and at position EU252 an arginine, an asparagine, an aspartic acid, a cysteine, a glutamic acid, a glutamine, a glycine, a histidine, a lysine, a methione, and at position EU252 an arginine, an asparagine, an aspartic acid, a cysteine,
  • modified FcRn-binding domain which comprises in addition to the substitutions at any one of the herein mentioned positions or combinations of positions, at position EU239 a lysine and/or at position EU270 a phenylalanine.
  • the antigen-binding molecules of the present invention are not particularly limited, as long as they include an antigen-binding domain having a binding activity specific to a target antigen and an FcRn-binding domain of the present invention.
  • Preferred antigen-binding domains comprise, for example, domains having an antigen-binding region of an antibody.
  • the antigen-binding region of an antibody comprises, for example, CDRs.
  • the antigen-binding region of an antibody may contain all six CDRs from the whole antibody, or one, two, or more CDRs.
  • the antigen-binding region of antibody comprise amino acid deletions, substitutions, additions, and/or insertions, or it may comprise a portion of CDR.
  • antigen-binding molecules of the present invention include antigen-binding molecules that have an antagonistic activity (antagonistic antigen-binding molecules), antigen-binding molecules that have an agonistic activity (agonistic antigen-binding molecule), and molecules having cytotoxicity.
  • the antigen-binding molecules are antagonistic antigen-binding molecules, in particular, antagonistic antigen-binding molecules that recognize an antigen such as a receptor or cytokine.
  • the antigen-binding molecule of the present invention is preferably an antibody.
  • the antibodies preferred in the context of the present invention include, for example, IgG antibodies.
  • the type of IgG is not particularly limited; thus, the IgG may belong to any isotype (subclass) such as IgG1, IgG2, IgG3, or IgG4.
  • IgG1, IgG2, IgG3, or IgG4 constant region gene polymorphisms (allotypes) are described in “Sequences of proteins of immunological interest, NIH Publication No. 91-3242”. These allotypes can also be used for constant region in this application.
  • both of the amino acids Asp-Glu-Leu (DEL) and Glu-Glu-Met (EEM) can be used for residues in 356-358 in EU numbering.
  • DEL Asp-Glu-Leu
  • EEM Glu-Glu-Met
  • gene polymorphisms allotypes are described in “Sequences of proteins of immunological interest, NIH Publication No. 91-3242”. These allotypes can also be used for constant region in this application.
  • the antigen-binding molecules of the present invention may include antibody constant region, and amino acid mutations may be introduced into the constant region.
  • Amino acid mutations to be introduced include, for example, those potentiate or impair the binding to Fcgamma receptor (Proc Natl Acad Sci USA. 2006 Mar. 14; 103(11): 4005-10), but are not limited to these examples.
  • an antigen-binding molecule of the present invention is an antibody
  • the antibody may be derived from any animal, such as a mouse, human, rat, rabbit, goat, or camel.
  • the antibody is a human antibody.
  • the antibody may be an altered antibody, for example, a chimeric antibody, and in particular, an altered antibody that comprises an amino acid substitution in the sequence of a humanized antibody, etc.
  • the category of antibodies contemplated by the present invention also include bispecific antibodies, antibody modification products linked with various molecules, and polypeptides that comprise antibody fragments (particularly immunogenic and/or immunoreactive antibody fragments).
  • the antigen-binding molecule is a monoclonal antibody.
  • Chimeric antibodies are antibodies prepared by combining sequences derived from different animals.
  • a chimeric antibody includes, for example, antibodies having heavy and light chain variable (V) regions from a mouse antibody and heavy and light chain constant (C) regions from a human antibody. Methods for generating chimeric antibodies are known.
  • V heavy and light chain variable
  • C heavy and light chain constant
  • Methods for generating chimeric antibodies are known.
  • a DNA encoding an antibody V region may be linked to a DNA encoding a human antibody C region; this can be inserted into an expression vector and introduced into a host to produce the chimeric antibody.
  • Humanized antibodies also referred to as reshaped human antibodies, are known in the art as antibodies in which complementarity determining regions (CDRs) of an antibody derived from a nonhuman mammal, for example, a mouse, are transplanted into the CDRs of a human antibody.
  • CDRs complementarity determining regions
  • Methods for identifying CDRs are known (Kabat et al., Sequence of Proteins of Immunological Interest (1987), National Institute of Health, Bethesda, Md.; Chothia et al., Nature (1989) 342: 877).
  • General genetic recombination technologies suitable for this purpose are also known (see European Patent Application EP 125023; and WO 96/02576).
  • Humanized antibodies can be produced by known methods, for example, the CDR of a mouse antibody can be determined, and a DNA encoding an antibody in which the CDR is linked to the framework region (FR) of a human antibody is obtained. Humanized antibodies can then be produced using a system that uses conventional expression vectors. Such DNAs can be synthesized by PCR, using as primers several oligonucleotides prepared to have portions that overlap with the end regions of both the CDR and FR (see the method described in WO 98/13388). Human antibody FRs linked via CDRs are selected such that the CDRs form a suitable antigen binding site.
  • amino acids in the FRs of an antibody variable region may be altered so that the CDRs of the reshaped human antibody can form a suitable antigen binding site (Sato et al., Cancer Res. (1993) 53: 10.01-6).
  • Amino acid residues in the FRs that can be altered include portions that directly bind to an antigen via non-covalent bonds (Amit et al., Science (1986) 233: 747-53), portions that influence or have an effect on the CDR structure (Chothia et al., J. Mol. Biol. (1987) 196: 901-17), and portions involved in VH-VL interactions (EP 239400).
  • the constant regions of these antibodies are preferably derived from human antibodies.
  • C-gamma1, C-gamma2, C-gamma3, and C-gamma4 can be used for the H chain, while C-kappa and C-lambda can be used for the L chain.
  • amino acid mutations may be introduced into the human antibody C region to enhance or lower the binding to Fc-gamma receptor or to improve antibody stability or productivity.
  • a chimeric antibody of the present invention preferably includes a variable region of an antibody derived from a nonhuman mammal and a constant region derived from a human antibody.
  • a humanized antibody preferably includes CDRs of an antibody derived from a nonhuman mammal and FRs and C regions derived from a human antibody.
  • the constant regions derived from human antibodies preferably include a human FcRn-binding region.
  • Such antibodies include, for example, IgGs (IgG1, IgG2, IgG3, and IgG4).
  • the constant regions used for the humanized antibodies of the present invention may be constant regions of antibodies of any isotype.
  • a constant region derived from human IgG1 is preferably used, though it is not limited thereto.
  • the FRs derived from a human antibody, which are used for the humanized antibodies are not particularly limited either, and may be derived from an antibody of any isotype.
  • bispecific antibody refers to an antibody that has, in the same antibody molecule, variable regions that recognize different epitopes.
  • a bispecific antibody may be an antibody that recognizes two or more different antigens, or an antibody that recognizes two or more different epitopes on a same antigen.
  • polypeptides including antibody fragments may be, for example, scFv-Fc (WO 2005/037989), dAb-Fc, and Fc fusion proteins.
  • Antibody fragments in such polypeptides can be for example Fab fragments, F(ab′)2 fragments, scFvs (Nat. Biotechnol. 2005 September; 23(9): 1126-36), domain antibodies (dAbs) (WO 2004/058821, WO 2003/002609)
  • Fc region can be used as a human FcRn-binding domain when a molecule includes an Fc region.
  • an FcRn-binding domain may be fused to these molecules.
  • antigen-binding molecules that are applicable to the present invention can be or can comprise antibody-like molecules (e.g. a fusion protein of an Fc region of the present invention with an antibody-like molecule).
  • An antibody-like molecule is a molecule that can exhibit functions by binding to a target molecule (Current Opinion in Biotechnology (2006) 17: 653-658; Current Opinion in Biotechnology (2007) 18: 1-10; Current Opinion in Structural Biology (1997) 7: 463-469; Protein Science (2006) 15: 14-27), and includes, for example, DARPins (WO 2002/020565), Affibody (WO 1995/001937), Avimer (WO 2004/044011; WO 2005/040229), and Adnectin (WO 2002/032925).
  • antibody-like molecules can bind to target molecules in a pH-dependent or calcium-dependent manner and/or have human FcRn-binding activity in the neutral pH range, it is possible to facilitate antigen uptake into cells by antigen-binding molecules, facilitate the reduction of plasma antigen concentration by administering antigen-binding molecules, and improve pharmacokinetics of the antigen-binding molecules, and increase the number of antigens to which a single antigen-binding molecule can bind.
  • the antigen-binding molecule can be a protein resulting from fusion between an FcRn-binding domain of the present invention and a receptor protein that binds to a target including a ligand, and includes, for example, TNFR-Fc fusion proteins, IL1R-Fc fusion proteins, VEGFR-Fc fusion proteins, and CTLA4-Fc fusion proteins (Nat Med. 2003, January; 9(1): 47-52; BioDrugs. (2006) 20(3): 151-60).
  • receptor-FcRn-binding domain fusion proteins bind to a target molecule including a ligand in a pH-dependent or calcium-dependent manner in addition to having FcRn-binding activity in the neutral pH range, it is possible to facilitate antigen uptake into cells by antigen-binding molecules, facilitate the reduction of plasma antigen concentration by administering antigen-binding molecules, and improve pharmacokinetics of the antigen-binding molecules, and increase the number of antigens to which a single antigen-binding molecule can bind.
  • a receptor protein is appropriately designed and modified so as to include a binding domain of the receptor protein to a target including a ligand. As referred to the examples hereinbefore (i.e.
  • a soluble receptor molecule comprising an extracellular domain of those receptor proteins that is required for binding to those targets including ligands is particularly preferred.
  • Such designed and modified receptor molecules are referred to as artificial receptors in the present invention. Methods for designing and modifying a receptor molecule to construct an artificial receptor molecule are known and indeed conventional in the art.
  • antibodies of the present invention can have modified sugar chains.
  • Antibodies with modified sugar chains include, for example, antibodies with modified glycosylation (WO 99/54342), antibodies that are deficient in fucose that is added to the sugar chain (WO 00/61739; WO 02/31140; WO 2006/067847; WO2006/067913), and antibodies having sugar chains with bisecting GlcNAc (WO 02/79255).
  • the human FcRn-binding activity of intact human IgG1 in the acidic pH range (pH 6.0) is KD 1.7 micromolar (microM), while in the neutral pH range the activity is almost undetectable.
  • the antigen-binding molecule of the present invention includes antigen-binding molecules whose human FcRn-binding activity in the acidic pH range is stronger than KD 1.7 micromolar and is identical or stronger in the neutral pH range than that of intact human IgG.
  • its binding activity for a pre-existing ADA in the neutral pH ranges is not significantly increased compared to intact IgG1.
  • the above KD values are determined by the method described in the Journal of Immunology (2009) 182: 7663-7671 (by immobilizing the antigen-binding molecule onto a chip and loading human FcRn as an analyte).
  • Dissociation constant can be used as a value of human FcRn-binding activity.
  • the human FcRn-binding activity of intact human IgG has little human FcRn-binding activity in the neutral pH range (pH 7.4). Accordingly, it is often difficult to calculate the activity as KD.
  • Methods for assessing whether the human FcRn-binding activity is higher than that of intact human IgG at pH 7.4 include assessment methods by comparing the intensities of Biacore response after loading analytes at the same concentration.
  • pH 7.0 can be used as the neutral pH range. Using pH 7.0 as a neutral pH can facilitate weak interaction between human FcRn and FcRn-binding domain.
  • a binding affinity may be assessed at any temperature from 10 degrees Celsius to 50 degrees Celsius. Preferably, a temperature ranging from 15 degrees Celsius to 40 degrees Celsius is employed in order to determine the binding affinity between human FcRn-binding domain and human FcRn. More preferably, any temperature ranging from 20 degrees Celsius to 35 degrees Celsius, like any one of 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, and 35 degrees C.
  • a temperature at 25 degrees C. described in EXAMPLE 5 of WO2011/122011 is one example for the embodiment of this invention.
  • an interaction between human FcRn and FcRn-binding domain can be measured at pH 7.0 and at 25 degrees C. as described in EXAMPLE 5 of WO2011/122011.
  • Binding affinity of antigen-binding molecule to human FcRn can be measured by Biacore as described in EXAMPLE 5 of WO2011/122011.
  • the binding affinity at neutral pH ranges is measured at pH 7.4, which is close to in vivo plasma (blood) pH.
  • pH 7.0 can be used as an alternative to pH 7.4 when it is difficult to assess the binding affinity between human FcRn-binding domain and human FcRn due its low affinity at pH 7.4.
  • the binding affinity at acidic pH ranges is measured at pH 6.0, which is close to the pH in early endosome in vivo.
  • a binding affinity between human FcRn-binding domain and human FcRn may be assessed at any temperature from 10 degrees C. to 50 degrees C.
  • a temperature from 15 degrees C. to 40 degrees C.
  • any temperature at from 20 degrees C. to 35 degrees C. is also employed in order to determine the binding affinity between human FcRn-binding domain and human FcRn.
  • a temperature at 25 degrees C. is described for example in Example 5 of WO2011/122011 and in the EXAMPLES of this invention.
  • an intact human IgG1, IgG2, IgG3 or IgG4 is preferably used as the reference intact human IgG to be compared with the antigen-binding molecules for their human FcRn binding activity or in vivo activity.
  • an antigen-binding molecule that comprises the same antigen-binding domain as the antigen-binding molecule of the interest and an intact human IgG Fc region as a human FcRn-binding domain is used as reference.
  • an intact human IgG1 is used as reference intact human IgG for comparing its human FcRn binding activity or in vivo activity with the human FcRn binding activity or in vivo activity of an antigen-binding molecule of the present invention.
  • Conditions used in the assay for the antigen-binding or human FcRn-binding activity other than pH can be appropriately selected by those skilled in the art, and the conditions are not particularly limited.
  • the conditions of using MES buffer at 37 degrees C. as described in WO 2009/125825 may be used to determine the activity.
  • Na-phosphate buffer at 25 degrees C. as described in Example 4 or 5 of WO2011/122011 may be used to determine the activity.
  • the antigen-binding activity and human FcRn-binding activity of antigen-binding molecule can be determined by methods known to those skilled in the art, for example, using Biacore (GE Healthcare) or such.
  • the activity of an antigen-binding molecule to bind to the soluble antigen can be determined by loading the antigen as an analyte onto a chip immobilized with the antigen-binding molecule.
  • the antigen is a membrane-type antigen
  • the activity of the antigen-binding molecule to bind to the membrane-type antigen can be determined by loading the antigen-binding molecule as an analyte onto an antigen-immobilized chip.
  • the human FcRn-binding activity of an antigen-binding molecule can be determined by loading human FcRn or the antigen-binding molecule as an analyte onto a chip immobilized with the antigen-binding molecule or human FcRn, respectively.
  • the present invention provides an antigen-binding molecule of the present invention that comprises an antigen-binding domain and a human Fc region having an increased FcRn-binding activity in the neutral pH ranges.
  • its binding activity for a pre-existing ADA in the neutral pH ranges is not significantly increased.
  • the FcRn-binding activity of such antigen-binding molecule in the neutral pH ranges is preferably stronger than KD 3.2 micromolar. More preferably, the FcRn-binding activity in the neutral pH range is stronger than 700 nanomolar, even more preferably stronger than 500 nanomolar and most preferably, stronger than 150 nanomolar.
  • the antigen-binding molecule has an increased human FcRn-binding activity in the neutral pH ranges and an antigen-binding activity that is lower in the acidic pH range than in the neutral pH range or that is lower at a low calcium concentration than at a high calcium concentration condition.
  • binding activity of such an antigen-binding molecule for a pre-existing ADA in the neutral pH ranges is not significantly increased.
  • the present invention also provides an antigen-binding molecule of the present invention that comprises an antigen-binding domain and a human FcRn-binding domain, wherein its human FcRn-binding activity is increased in the neutral pH ranges, further wherein the human FcRn-binding activity in the neutral pH ranges is 28-fold stronger than that of an intact human IgG, more preferably, the human FcRn-binding activity in the neutral pH ranges is 38-fold stronger than that of an intact human IgG.
  • binding activity of such an antigen-binding molecule for a pre-existing ADA in the neutral pH ranges is not significantly increased.
  • the antigen-binding molecule of the present invention with an increased FcRn-binding activity in the neutral pH ranges Preferably a binding activity for a pre-existing ADA in the neutral pH ranges that is not significantly increased preferably, has human FcRn-binding activity at pH 7.0 and at 25 degrees C. which is 28-fold stronger, preferably 38-fold stronger, than intact human IgG than intact human IgG.
  • the human FcRn-binding activity of the antigen-binding molecule with an increased FcRn binding activity at pH 7.0 and at 25 degrees C. is preferably stronger than KD 3.2 micromolar. More preferably, the FcRn-binding activity in at pH 7.0 and at 25 degrees Celsius is stronger than 700 nanomolar, more preferably stronger than 500 nanomolar and most preferably, stronger than 150 nanomolar.
  • the present invention provides an antigen-binding molecule of the present invention, comprising an antigen-binding domain and a human Fc region of the present invention, with an increased FcRn-binding activity in the acidic pH ranges and a binding activity for a pre-existing ADA in the neutral pH ranges that is not significantly increased.
  • the present invention also provides an antigen-binding molecule of the present invention comprising an antigen-binding domain and a human FcRn-binding domain having an increased human FcRn-binding activity in the acidic pH range and a binding activity for a pre-existing ADA in the neutral pH ranges that is not significantly increased as compared to the binding activity for a pre-existing ADA of an intact IgG, wherein the human FcRn-binding activity in the acidic pH ranges is in the range of about 2-fold to about 100-fold stronger than the human FcRn-binding activity of an intact human IgG.
  • the human FcRn-binding activity of antigen-binding molecule of the present invention in the acidic pH ranges is at least 10-fold stronger than the FcRn-binding activity of an intact human IgG, more preferably, the human FcRn-binding activity in the acidic pH ranges is at least 20-fold stronger than that of an intact human IgG.
  • the antigen-binding molecule of the present invention with an increased FcRn-binding activity in the acidic pH ranges whereby its binding activity for a pre-existing ADA in the neutral pH ranges is not significantly increased has human FcRn-binding activity at pH 6.0 and at 25 degrees C. which is 10-fold stronger, preferably 20-fold stronger, than intact human IgG.
  • the antigen-binding molecules of the present invention may have an increased FcRn-binding activity in the neutral pH ranges as well as an antigen-binding activity in the acidic pH range that is lower than the antigen-binding activity in the neutral pH range or an antigen-binding activity at a low calcium concentration that is lower than the antigen-binding activity at a high calcium concentration condition.
  • Specific examples of such antigen-binding molecules include those that have a higher binding activity for human FcRn at pH 7.4 than an intact Ig, and whose antigen-binding activity is lower at pH 5.8 than at pH 7.4 which are presumed to be the in vivo pH of the early endosome and plasma, respectively.
  • An antigen-binding molecule whose antigen-binding activity is lower at pH 5.8 than at pH 7.4 can also be referred to as an antigen-binding molecule whose antigen-binding activity is stronger at pH 7.4 than at pH 5.8.
  • the value of KD (pH 5.8)/KD (pH 7.4) which is a ratio of dissociation constant (KD) against an antigen at pH 5.8 and pH 7.4, is 1.5, 2, 3, 4, 5, 10, 15, 20, 50, 70, 80, 100, 500, 1000 or 10,000 preferably 2 or greater, more preferably 10 or greater, and still more preferably 40 or greater.
  • the upper limit of the KD (pH 5.8)/KD (pH 7.4) value is not particularly limited, and may be any value, for example, 400, 1,000, or 10,000, as long as production is possible using the technologies of those skilled in the art.
  • antigen-binding molecules of the present invention that have an increased FcRn-binding activity in the acidic pH ranges, as well as a lower antigen-binding activity in the acidic pH range than that in the neutral pH range or a lower antigen-binding activity at a low calcium concentration than that at a high calcium concentration.
  • binding activity of such an antigen-binding molecule for a pre-existing ADA in the neutral pH ranges is not significantly increased.
  • antigen-binding molecules include those that have a higher binding activity for human FcRn at pH 5.8 to pH 6.0 than an IgG, which is presumed to be the in vivo pH of the early endosome and whose antigen-binding activity is lower at pH 5.8 than at pH 7.4.
  • An antigen-binding molecule whose antigen-binding activity is lower at pH 5.8 than at pH 7.4 can also be referred to as an antigen-binding molecule whose antigen-binding activity is weaker at pH 5.8 than at pH 7.4.
  • an antigen-binding molecule having an increased binding activity for FcRn in the acidic pH ranges has stronger FcRn-binding activity than intact human IgG in the neutral pH range.
  • modified FcRn-binding domains of the present invention are applicable to any antigen-binding molecules, regardless of the type of target antigen.
  • An antigen-binding molecule of the present invention may have other properties.
  • it may be an agonistic or antagonistic antigen-binding molecule, provided that it has a) the requisite increased human FcRn-binding activity neutral pH ranges, or b) an increased human FcRn-binding activity for in the acidic ranges and its binding activity for a pre-existing ADA is not significantly increased.
  • the antigen-binding activity of such an antigen-binding molecule is lower in the acidic pH range than in the neutral pH range.
  • Preferred antigen-binding molecules of the present invention include, for example, antagonistic antigen-binding molecules.
  • Such an antagonistic antigen-binding molecule is typically an antigen-binding molecule that inhibits receptor-mediated intracellular signaling by blocking the binding between ligand (agonist) and receptor.
  • an antigen-binding molecule of the present invention may recognize any antigen.
  • Specific antigens recognized by an antigen-binding molecule of the present invention include, for example, the above-described receptor proteins (membrane-bound receptors and soluble receptors), membrane antigens such as cell-surface markers, and soluble antigens such as cytokines
  • Such antigens include, for example, the antigens described below.
  • Antigen-binding molecules of the present invention comprising an antigen-binding domain can utilize a difference of pH as an environmental difference between plasma and endosome for differential binding affinity of an antigen binding molecule to an antigen at plasma and endosome (strong binding at plasma and weak binding at endosome). Since environmental difference between plasma and endosome is not limited to a difference of pH, pH dependent binding property on binding of an antigen-binding molecule to an antigen can be substituted by utilizing other factors whose concentration is different within the plasma and the endosome, such as for example the ionized calcium concentration. Such factor may also be used to generate an antibody that binds to the antigen within plasma but dissociates the antigen within endosome.
  • the present invention also includes an antigen-binding molecule comprising a human FcRn-binding domain, whose human FcRn-binding activity is increased in the neutral pH ranges and whose antigen-binding activity in the endosome is lower as compared to the plasma.
  • an antigen-binding molecule comprising a human FcRn-binding domain, whose human FcRn-binding activity is increased in the neutral pH ranges and whose antigen-binding activity in the endosome is lower as compared to the plasma.
  • the binding activity of these antigen-binding molecules in the neutral pH ranges for a pre-existing ADA is not significantly increased.
  • the human FcRn-binding activity of such an antigen-binding molecule is in the plasma stronger than that of intact human IgG, and further the antigen-binding domain of such an antigen-binding molecule has a lower affinity for the antigen inside the endosome than in the plasma.
  • the antigen-binding domain is an antigen-binding domain whose antigen-binding activity in the acidic pH range is lower than that in the neutral pH range (pH-dependent antigen-binding domain) or an antigen-binding domain whose antigen-binding activity is lower with a low calcium concentration than under a high calcium concentration condition (calcium-concentration-dependent antigen-binding domain).
  • the present invention also includes an antigen-binding molecule with a human FcRn-binding domain, which has an increased human FcRn-binding activity in the acidic pH ranges, and said antigen-binding molecule further comprises an antigen-binding domain which has a lower affinity for the antigen inside the endosome than in the plasma, such that the human FcRn-binding activity of the antigen-binding molecule in the endosome is stronger than that of intact human IgG, and the antigen-binding activity of the antigen-binding molecule in the endosome is stronger than in the plasma.
  • the binding activity of these antigen-binding molecules in the neutral pH ranges for a pre-existing ADA is not significantly increased.
  • the antigen-binding domain is an antigen-binding domain whose antigen-binding activity in the acidic pH range is lower than that in the neutral pH range (pH-dependent antigen-binding domain) or an antigen-binding domain whose antigen-binding activity is lower with a low calcium concentration than under a high calcium concentration condition (calcium-concentration-dependent antigen-binding domain).
  • the antigen-binding molecules of the present invention facilitate antigen uptake into cells, in particular when the antigen-binding molecules of the present invention comprising an antigen-binding domain that is a pH-dependent antigen-binding domain or a calcium-concentration-dependent antigen-binding domain.
  • the antigen-binding molecules are easily dissociated from the antigen in the endosome, and then released to the outside of the cell by binding to human FcRn.
  • the antigen-binding molecules of the present invention are presumed to bind easily to an antigen in the plasma again.
  • the antigen-binding molecule of the present invention is a neutralizing antigen-binding molecule, reduction of the plasma antigen concentration can be facilitated by administering the molecule.
  • the antigen-binding domain of the antigen-binding molecule has a decreased affinity for the antigen at an acidic pH or at low calcium ion concentration. More preferably, the antigen-binding domain is a pH-dependent antigen-binding domain or a ionized calcium concentration dependent antigen-binding domain described herein.
  • the antigen-binding molecule of the present invention comprises preferably a pH-dependent antigen-binding domain whose antigen-binding activity in the acidic pH range is lower than that in the neutral pH range.
  • Said antigen-binding molecule has preferably a lower antigen-binding activity in the acidic pH range than in the neutral pH range.
  • the binding activity ratio is not limited, provided that the antigen-binding activity is lower in the acidic pH range than in the neutral pH range.
  • the antigen-binding molecules of the present invention include antigen-binding molecules whose antigen-binding activity at pH 7.4 is twice or higher than that at pH 5.8, preferably the antigen-binding activity at pH 7.4 is ten times or higher than that at pH 5.8.
  • the antigen-binding molecules of the present invention include antigen-binding molecules whose antigen-binding activity at pH 7.4 is 40 times or higher than that at pH 5.8.
  • antigen-binding molecules of the present invention include the embodiments described in WO 2009/125825.
  • the antigen-binding molecule of the present invention comprising a pH-dependent antigen-binding domain has an antigen-binding activity at pH 5.8 that is lower than that at pH 7.4, wherein the value of KD (pH5.8)/KD (pH7.4), which is a ratio of KD for the antigen at pH 5.8 and that at pH 7.4, is preferably 2 or greater, more preferably 10 or greater, and still more preferably 40 or greater.
  • the upper limit of the KD (pH5.8)/KD (pH7.4) value is not particularly limited, and may be any value, for example, 400, 1,000, or 10,000, as long as production is possible using the technologies of those skilled in the art.
  • the antigen-binding molecule of the present invention whose antigen-binding activity at pH 5.8 is lower than that at pH 7.4, has a value of KD (pH5.8)/KD (pH7.4), which is a ratio of the KD for the antigen at pH 5.8 and the KD for the antigen at pH 7.4, that is 2 or greater, more preferably 5 or greater, even more preferably 10 or greater, and still more preferably 30 or greater.
  • the upper limit of the KD (pH5.8)/KD (pH7.4) value is not particularly limited, and may be any value, for example, 50, 100, or 200, provided that the production is possible using the technologies of those skilled in the art.
  • Conditions other than the pH at which the antigen-binding activity, binding activity for a pre-existing ADA and human FcRn-binding activity are measured can be appropriately selected by those skilled in the art, and such conditions are not particularly limited; however, the measurements can be carried out, for example, under conditions of MES buffer and at 37 degrees C., as described in the Examples.
  • the antigen-binding activity of an antigen-binding molecule can be determined by methods known to those skilled in the art, for example, using Biacore T100 (GE Healthcare) or the like, as described in the Examples.
  • Methods for reducing (impairing) the antigen-binding activity of an antigen-binding molecule in the acidic pH range to less than that the antigen-binding activity in the neutral pH range are not particularly limited and suitable methods are known to the skilled in the art.
  • WO 2009/125825 describes methods for reducing (impairing) the antigen-binding activity in the acidic pH range to less than that in the neutral pH range by substituting histidine for an amino acid in the antigen-binding domain or inserting histidine into the antigen-binding domain.
  • an antibody can be conferred with a pH-dependent antigen-binding activity by substituting histidine for an amino acid in the antibody (FEBS Letter (1992) 309(1): 85-88).
  • Other suitable methods include methods for substituting non-natural amino acids for amino acids in the antigen-binding domain or inserting non-natural amino acids into the antigen-binding domain.
  • pKa can be artificially adjusted by using non-natural amino acids (Angew. Chem. Int. Ed. 2005, 44, 34; Chem Soc Rev. 2004 Sep. 10, 33 (7): 422-30; Amino Acids. (1999) 16(3-4): 345-79). Any non-natural amino acid may be used in context of the present invention. In fact, it is possible to use non-natural amino acids known to those skilled in the art.
  • the antigen-binding molecule of the present invention comprising an antigen-binding domain with an antigen-binding activity that is lower in the acidic pH range than that in the neutral pH range, includes antigen-binding molecules in which at least one amino acid in the antigen-binding molecule is replaced with histidine or a non-natural amino acid, and/or in which at least one histidine or a non-natural amino acid has been inserted.
  • the site into which the histidine or non-natural amino acid mutation is introduced is not particularly limited and may be any site deemed suitable by those of skilled in the art, provided that the resultant antigen-binding activity in the acidic pH range is weaker than that in the neutral pH range (the KD (in the acidic pH range)/KD (in the neutral pH range) value is greater or the kd (in the acidic pH range)/kd (in the neutral pH range) value is greater) as compared to before substitution.
  • Examples include variable regions and CDRs of an antibody in the case the antigen-binding molecule is an antibody.
  • the number of amino acids to be replaced with histidine or non-natural amino acid and the number of amino acids to be inserted can be appropriately determined by those skilled in the art.
  • One amino acid may be replaced with histidine or non-natural amino acid, or one amino acid may be inserted, or two or more amino acids may be replaced with histidine or non-natural amino acids, or two or more amino acids may be inserted.
  • substitutions of histidine or non-natural amino acid or insertion of histidine or of non-natural amino acid may also be simultaneously carried out.
  • Substitutions of histidine or non-natural amino acid or insertion of histidine or of non-natural amino acid may be carried out at random using a method such as histidine scanning, which uses histidine instead of alanine in alanine scanning which is known to those skilled in the art.
  • Antigen-binding molecules whose KD (pH5.8)/KD (pH7.4) or kd (pH5.8)/kd (pH7.4) is increased as compared to before mutation can be selected from antigen-binding molecules into which histidine or non-natural amino acid mutation has been introduced at random.
  • the binding activity of the antigen-binding domain at neutral pH is maintained.
  • the antigen-binding activity of an antigen-binding molecule before histidine or non-natural amino acid mutation is set as 100%
  • the antigen-binding activity of the antigen-binding molecule at pH7.4 after histidine or non-natural amino acid mutation is at least 10% or more, preferably 50% or more, more preferably 80% or more, and still more preferably 90% or more.
  • the antigen-binding activity at pH 7.4 after histidine or non-natural amino acid mutation may be stronger than the antigen-binding activity at pH 7.4 before histidine or non-natural amino acid mutation.
  • the antigen-binding activity of the antigen-binding molecule may be adjusted by introducing substitution, deletion, addition, and/or insertion and such of one or more amino acids into the antigen-binding molecule so that the antigen-binding activity becomes equivalent to that before histidine substitution or insertion.
  • possible sites of histidine or non-natural amino acid substitution include, for example, CDR sequences and sequences responsible for the CDR structure of an antibody, including, for example, the sites described in WO 2009/125825.
  • the present invention provides antigen-binding molecules having substitution of histidine or a non-natural amino acid for at least one amino acid at one of the following sites
  • Heavy chain H27, H31, H32, H33, H35, H50, H58, H59, H61, H62, H63, H64, H65, H99, H100b, and H102
  • Light chain L24, L27, L28, L32, L53, L54, L56, L90, L92, and L94
  • H32, H61, L53, L90, and L94 of these alteration sites are presumed to be highly general alteration sites.
  • the amino acid positions are shown according to Kabat numbering (Kabat et al., Sequences of Immunological Interest. 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991)).
  • the Kabat numbering system is generally used when referring to a residue in the variable domain (approximately residues 1-107 of the light chain and residues 1-113 of the heavy chain).
  • Specifically preferred combinations of sites for histidine or non-natural amino acid substitutions include, for example, the combination of H27, H31, and H35; the combination of H27, H31, H32, H35, H58, H62, and H102; the combination of L32 and L53; and the combination of L28, L32, and L53.
  • preferred combinations of substitutions sites in the heavy and light chains include, for example, the combination of H27, H31, L32, and L53.
  • preferred alteration sites include but are not particularly limited to the following:
  • Heavy chain H27, H31, H32, H35, H50, H58, H61, H62, H63, H64, H65, H100b, and H102
  • Light chain L24, L27, L28, L32, L53, L56, L90, L92, and L94
  • Specifically preferred combinations of sites for histidine or non-natural amino acid substitution include, for example, the combination of H27, H31, and H35; the combination of H27, H31, H32, H35, H58, H62, and H102; the combination of L32 and L53; and the combination of L28, L32, and L53.
  • preferred combinations of substitution sites in the heavy and light chains include, for example, the combination of H27, H31, L32, and L53.
  • Histidine or non-natural amino acids can be substituted at one or more of the positions mentioned above.
  • the antigen-binding molecule of the present invention may comprise an antibody constant region that was altered so that the antigen-binding activity at pH 5.8 is lower than that at pH 7.4.
  • Methods for altering antibody constant regions contained in the antigen-binding molecules are known and indeed conventional to the skilled in the art. Specific examples of antibody constant regions after alteration include the constant regions described in the Examples in WO 2009/125825 (SEQ ID NOs: 11, 12, 13, and 14).
  • methods for altering an antibody constant region include, for example, methods for assessing various constant region isotypes (IgG1, IgG2, IgG3, and IgG4) and selecting isotypes that reduce the antigen-binding activity in the acidic pH range (increase the dissociation rate in the acidic pH range) are known. Such methods also include methods for reducing the antigen-binding activity in the acidic pH range (increasing the dissociation rate in the acidic pH range) by introducing amino acid substitutions into the amino acid sequences of wild-type isotypes (amino acid sequences of wild type IgG1, IgG2, IgG3, or IgG4).
  • the sequence of hinge region in the antibody constant region is considerably different among isotypes (IgG1, IgG2, IgG3, and IgG4), and the difference in the hinge region amino acid sequence has a great impact on the antigen-binding activity.
  • IgG1, IgG2, IgG3, and IgG4 isotypes
  • the difference in the hinge region amino acid sequence has a great impact on the antigen-binding activity.
  • preferred amino acid substitution sites in the amino acid sequences of wild type isotypes are presumed to be within the hinge region.
  • the above-described methods can be used to produce antigen-binding molecules whose antigen-binding activity in the acidic pH range is reduced (weakened) to less than that in the neutral pH range (antigen-binding molecules that bind in a pH-dependent manner) by amino acid substitution or insertion from antigen-binding molecules that do not have such property.
  • Other methods include methods for directly obtaining antigen-binding molecules having the above-described property.
  • antibodies having a desired property of interest may be directly selected by screening using the pH-dependent antigen binding as an indicator from antibodies obtained by immunizing animals (mice, rats, hamsters, rabbits, human immunoglobulin-transgenic mice, human immunoglobulin-transgenic rats, human immunoglobulin-transgenic rabbits, llamas, camels, etc.) with an antigen.
  • Antibodies can be generated by hybridoma technology or B-cell cloning technology (Bernasconi et al, Science (2002) 298, 2199-2202; WO2008/081008) which are methods known to those skilled in the art, but not limited thereto.
  • antibodies that have the property of interest may be directly selected by screening using the pH-dependent antigen binding as an indicator from a library of presenting antigen-binding domain in vitro.
  • library includes human naive library, immunized library from non-human animal and human, semi-synthetic library and synthetic library which are libraries known to those skilled in the art (Methods Mol Biol. 2002; 178: 87-100; J Immunol Methods. 2004 June; 289(1-2): 65-80; and Expert Opin Biol Ther. 2007 May; 7(5): 763-79), but not limited thereto.
  • the methods are not particularly limited to these examples.
  • the antigen-binding molecule of the present invention comprises a calcium-ion dependent antigen-binding domain.
  • the antigen-binding activity of such an antigen-binding molecule depends of the calcium concentration, whereby the antigen-binding activity at a low calcium concentration is lower than that at a high calcium concentration.
  • the antigen-binding activity includes the antigen-binding activity at an ionized calcium concentration of 0.5 to 10 micromolar. More preferable ionized calcium concentrations include the ionized calcium concentration in the early endosome in vivo. Specifically, the antigen-binding activity includes the activity at 1 to 5 micromolar. Meanwhile, the antigen-binding activity of an antigen-binding molecule at a high calcium concentration is not particularly limited, provided that it is the antigen-binding activity at an ionized calcium concentration of 100 micromolar to 10 mM. Preferably, the antigen-binding activity includes the antigen-binding activity at an ionized calcium concentration of 200 micromolar to 5 mM. Preferably, a low calcium concentration is an ionized calcium concentration of 0.1 to 30 micromolar, and a high calcium concentration is an ionized calcium concentration of 100 micromolar to 10 mM.
  • the low calcium concentration is an intraendosomal concentration of ionized calcium
  • the high calcium concentration is a plasma concentration of ionized calcium
  • the antigen-binding molecules comprising said calcium-dependent antigen-binding domain include antigen-binding molecules whose antigen-binding activity at the ionized calcium concentration in the early endosome in vivo (a low calcium concentration of such as 1 to 5 micromolar) is lower than that at the ionized calcium concentration in plasma in vivo (a high calcium concentration of such as 0.5 to 2.5 mM).
  • the antigen-binding activity of an antigen-binding molecule whose antigen-binding activity at a low calcium concentration is lower than that at a high calcium concentration there is no limitation on this difference in the antigen-binding activity, provided that the antigen-binding activity at a low calcium concentration is lower than that at a high calcium concentration. It is even acceptable that the antigen-binding activity of an antigen-binding molecule is only slightly lower under a low calcium concentration condition.
  • the value of KD (low Ca)/KD (high Ca), which is the KD ratio between low and high calcium concentration, is 2 or more, preferably the value of KD (low Ca)/KD (high Ca) is 10 or more, and more preferably the value of KD (low Ca)/KD (high Ca) is 40 or more.
  • the upper limit of the KD (low Ca)/KD (high Ca) value is not particularly limited, and may be any value such as 400, 1,000, and 10,000 provided that it can be produced by techniques known to those skilled in the art.
  • the value of kd (low Ca)/kd (high Ca), which is the ratio of kd for an antigen between a low calcium concentration condition and pH 7.4, is 2 or more, preferably the value of kd (low Ca)/kd (high Ca) is 5 or more, more preferably the value of kd (low Ca)/kd (high Ca) is 10 or more, and still more preferably the value of kd (low Ca)/kd (high Ca) is 30 or more.
  • the upper limit of kd (low Ca)/kd (high Ca) value is not particularly limited, and may be any value such as 50, 100, and 200 as long as it can be produced by techniques known to those skilled in the art.
  • the antigen-binding activity of an antigen-binding molecule can be determined by methods known to those skilled in the art. Appropriate conditions besides ionized calcium concentration can be selected by those skilled in the art.
  • the antigen-binding activity of an antigen-binding molecule can be assessed by using KD (dissociation constant), apparent KD (apparent dissociation constant), dissociation rate kd (dissociation rate), apparent kd (apparent dissociation: apparent dissociation rate), or the like. They can be determined by methods known to those skilled in the art, for example, using Biacore (GE Healthcare), Scatchard plot, FACS, or such.
  • Antigen-binding molecules to be screened by the screening method of the present invention may be any antigen-binding molecules. It is possible to screen, for example, antigen-binding molecules having a natural sequence or antigen-binding molecules having an amino acid sequence with a substitution. Antigen-binding molecules comprising a calcium-ion dependent antigen-binding domain to be screened by the screening method of the present invention may be prepared by any methods.
  • preexisting antibodies for example, preexisting antibodies, preexisting libraries (phage libraries, etc.), and antibodies and libraries prepared from B cells of immunized animals or hybridomas prepared by immunizing animals, antibodies or libraries obtained by introducing amino acids capable of chelating calcium (for example, aspartic acid or glutamic acid) or non-natural amino acid mutations into such antibodies or libraries (libraries with high content of non-natural amino acids or amino acids capable of chelating calcium (for example, aspartic acid or glutamic acid), libraries introduced with non-natural amino acid mutations or mutations with amino acids capable of chelating calcium (for example, aspartic acid or glutamic acid) at specific sites, or such), or the like.
  • amino acids capable of chelating calcium for example, aspartic acid or glutamic acid
  • non-natural amino acid mutations for example, aspartic acid or glutamic acid
  • An antigen-binding molecule whose antigen-binding activity under a low calcium concentration condition is lower than that under a high calcium concentration condition can be readily screened, identified and isolated using methods conventional in the art (see e.g. PCT application no. PCT/JP2011/077619. Examples of such screening methods include the step of assaying for an antigen-binding molecule having at least one function selected from:
  • the present invention provides methods of screening for an antigen-binding molecule comprising a calcium-ion dependent antigen-binding domain, which comprises the steps of:
  • a method for producing an antigen-binding molecule with a calcium-ion dependent antigen-binding domain is for example a method comprising the steps of:
  • Another method of producing an antigen-binding molecule with a calcium-ion dependent antigen-binding domain is the method comprising the steps of:
  • step (b) obtaining an antigen-binding molecule that bound to the antigen in step (a);
  • step (c) allowing the antigen-binding molecule obtained in step (b) to stand under a low calcium concentration condition;
  • step (d) obtaining an antigen-binding molecule whose antigen-binding activity in step (c) is lower than the activity for the selection in step (b);
  • step (e) obtaining a gene encoding the antigen-binding molecule obtained in step (d);
  • step (f) producing the antigen-binding molecule using the gene obtained in step (e).
  • Steps (a) to (e) may be repeated two or more times.
  • the present invention provides the methods further comprising the step of repeating steps (a) to (e) two or more times in the above-described methods.
  • the number of repetitions of steps (a) to (e) is not particularly limited; however, the number is generally ten or less.
  • Antigen-binding molecules that are used in the production methods of the present invention may be prepared by any conventional method.
  • pre-existing antibodies, pre-existing libraries (phage libraries and the like) antibodies and libraries that are prepared from hybridomas obtained by immunizing animals or from B cells of immunized animals, antibodies and libraries prepared by introducing histidine or non-natural amino acid mutations into the above-described antibodies and libraries (libraries with high content of histidine or non-natural amino acid, libraries introduced with histidine or non-natural amino acid at specific sites, and the like), and such.
  • Antigens that are recognized by antigen-binding molecules of the present invention are not particularly limited. Such antigen-binding molecules of the present invention may recognize any antigen. Specific examples of an antigen that is recognized by the antigen-binding molecule of the present invention include but are not limited to: 17-IA, 4-1 BB, 4Dc, 6-keto-PGF1a, 8-iso-PGF2a, 8-oxo-dG, A1 Adenosine Receptor, A33, ACE, ACE-2, Activin, Activin A, Activin AB, Activin B, Activin C, Activin RIA, Activin RIA ALK-2, Activin RIB ALK-4, Activin RIIA, Activin RIIB, ADAM, ADAM10, ADAM12, ADAM15, ADAM17/TACE, ADAMS, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, Addressins,
  • amyloid immunoglobulin light chain variable region Androgen, ANG, angiotensinogen, Angiopoietin ligand-2, anti-Id, antithrombin III, Anthrax, APAF-1, APE, APJ, apo A1, apo serum amyloid A, Apo-SAA, APP, APRIL, AR, ARC, ART, Artemin, ASPARTIC, Atrial natriuretic factor, Atrial natriuretic peptide, atrial natriuretic peptides A, atrial natriuretic peptides B, atrial natriuretic peptides C, av/b3 integrin, Axl, B7-1, B7-2, B7-H, BACE, BACE-1, Bacillus anthracis protective antigen, Bad, BAFF, BAFF-R, Bag-1, BAK, Bax, BCA-1, BCAM, Bcl, BCMA, BDNF, b-
  • HGF Hemopoietic growth factor
  • Hep B gp120 Heparanase
  • heparin cofactor II hepatic growth factor
  • Bacillus anthracis protective antigen Hepatitis C virus E2 glycoprotein, Hepatitis E, Hepcidin, Her1, Her2/neu (ErbB-2), Her3 (ErbB-3), Her4 (ErbB-4), herpes simplex virus (HSV) gB glycoprotein, HGF, HGFA, High molecular weight melanoma-associated antigen (HMW-MAA), HIV envelope proteins such as GP120, HIV MIB gp120 V3 loop, HLA, HLA-DR, HM1.24, HMFG PEM, HMGB-1, HRG, Hrk, HSP47, Hsp90,
  • Antigen binding molecules described in present invention are capable of reducing total antigen concentration of the above-described antigens in plasma. Antigen binding molecules described in present invention are also capable of eliminating virus, bacteria, and fungus from plasma by binding to structural components of virus, bacteria and fungus. Particularly, F protein of RSV, Staphylococcal lipoteichoic acid, Clostridium difficile toxin, Shiga like toxin II, Bacillus anthracis protective antigen and Hepatitis C virus E2 glycoprotein can be used as a structural components of virus, bacteria and fungus.
  • the present invention also provides many uses of the antigen-binding molecules of the present invention as described above.
  • the present invention provides the use of the modified antigen-binding molecules of the present invention for improving the antigen-binding molecule-mediated antigen uptake into cells. Furthermore, the present invention also provides methods for improving antigen-binding molecule-mediated antigen uptake into cells comprising altering an antigen-binding molecule comprising a parent FcRn-binding domain, by substituting an amino acid in the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and thereby increasing the FcRn-binding activity at neutral pH as compared to an antigen-binding molecule having an intact FcRn-binding domain.
  • the term “antigen uptake into cells” mediated by an antigen-binding molecule means that antigens are taken up into cells by endocytosis.
  • the term “facilitate the uptake into cells” means that the rate of intracellular uptake of antigen-binding molecule bound to an antigen in plasma is enhanced, and/or the quantity of recycling of uptaken antigen to the plasma is reduced.
  • the rate of uptake into cells is facilitated as compared to the antigen-binding molecule before the modification of the FcRn-binding domain and thus before increasing the human FcRn-binding activity of the antigen-binding molecule in the neutral pH range, or before increasing the human FcRn-binding activity and reducing the antigen-binding activity (binding ability) of the antigen-binding molecule in the acidic pH range to less than its antigen-binding activity in the neutral pH range.
  • the rate is improved preferably as compared to intact IgG, and more preferably as compared to intact human IgG.
  • whether antigen uptake into cells is facilitated by an antigen-binding molecule can be assessed based on an increase in the rate of antigen uptake into cells.
  • the rate of antigen uptake into cells can be calculated, for example, by monitoring over time reduction in the antigen concentration in the culture medium containing human FcRn-expressing cells after adding the antigen and antigen-binding molecule to the medium, or monitoring over time the amount of antigen uptake into human FcRn-expressing cells.
  • the rate of antigen elimination from the plasma can be enhanced by administering antigen-binding molecules of the present invention.
  • antigen-binding molecule-mediated antigen uptake into cells is facilitated can also be assessed, for example, by testing whether the rate of antigen elimination from the plasma is accelerated or whether the total antigen concentration in plasma is reduced by administering an antigen-binding molecule of the present invention.
  • total antigen concentration in plasma means the sum of antigen-binding molecule bound antigen and non-bound antigen concentration, or “free antigen concentration in plasma” which is antigen-binding molecule non-bound antigen concentration.
  • Various methods to measure “total antigen concentration in plasma” and “free antigen concentration in plasma” are well known in the art as described hereinafter.
  • the present invention also provides use of the antigen-binding molecule of the present invention for increasing the total number of antigens to which a single antigen-binding molecule can bind before its degradation.
  • the present invention also provides methods for increasing the number of antigens to which a single antigen-binding molecule can bind, by using an antigen-binding molecule of the present invention.
  • the present invention provides methods for increasing the total number of antigens to which a single antigen-binding molecule can bind, by substituting an amino acid in the parent FcRn-binding domain of said antigen-binding molecule at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and thereby increasing the FcRn-binding activity at neutral pH as compared to an antigen-binding molecule having an intact FcRn-binding domain.
  • a “conventional antibody” can usually bind only one or two antigens before it is degraded in the endosome.
  • An antigen-binding molecule of the present invention can increase the number of cycles achieved until the antigen-binding molecule is degraded, whereby each cycle consists of: binding of an antigen to the antigen-binding molecule in plasma, intracellular uptake of the antigen-binding molecule bound to the antigen, and dissociation from the antigen in the endosome, followed by return of the antigen-binding molecule to the plasma.
  • the number of cycles is increased as compared to the antigen-binding molecule before the modification of the FcRn-binding domain and thus before increasing the human FcRn-binding activity of the antigen-binding molecule in the neutral pH or acidic range, or before increasing the human FcRn-binding activity and reducing the antigen-binding activity (binding ability) of the antigen-binding molecule in the acidic pH range to less than its antigen-binding activity in the neutral pH range.
  • whether the number of cycles is increased can be assessed by testing whether the above-described “intracellular uptake is facilitated” or whether the “pharmacokinetics is improved” as described below.
  • the present invention also provides for the use of the antigen-binding molecules of the present invention for improving the antigen-removal from the blood in mammals, i.e. in humans.
  • the present invention provides the use of the antigen-binding molecule of the present invention for reducing the plasma concentration of a specific antigen, wherein the antigen-binding molecule comprises an antigen-binding domain which can bind said antigen.
  • the present invention also provides a method for reducing the plasma concentration of a specific antigen, wherein the antigen-binding molecule comprises an antigen-binding domain which can bind said antigen, by substituting an amino acid in a parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and thereby increasing the FcRn-binding activity at neutral pH as compared to an antigen-binding molecule having an intact FcRn-binding domain.
  • the present invention also provides the use of the antigen-binding molecules of the present invention for facilitating the extracellular release of antigen-free antigen-binding molecule taken up into cells in an antigen-bound form. More specifically, the present invention provides methods for facilitating the extracellular release of antigen-free antigen-binding molecule taken up into cells in an antigen-bound form without significantly increasing the binding activity for a pre-existing ADA at neutral pH compared to parent antibody, by substituting an amino acid in the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and thereby increasing the FcRn-binding activity at neutral pH as compared to an antigen-binding molecule having an intact FcRn-binding domain
  • the “extracellular release of antigen-free antigen-binding molecule taken up into cells in an antigen-bound form” does not necessarily mean that all of the antigen-binding molecules bound to antigen taken up into cells are released in an antigen-free form outside of the cell. It is acceptable that the proportion of antigen-binding molecules released in an antigen-free form to the outside of the cell is increased as compared to before the modification of the FcRn-binding domain and thus before reducing the antigen-binding activity of the antigen-binding molecule in the acidic pH range to less than that in the neutral pH range and increasing the human FcRn-binding activity in the neutral pH range.
  • the antigen-binding molecule released to the outside of the cell preferably retains the antigen-binding activity.
  • the present invention also provides the use of an FcRn-binding domain of the present invention for increasing the ability of the antigen-binding molecule to eliminate plasma antigen.
  • “methods for increasing the ability to eliminate plasma antigen” is synonymous to “methods for augmenting the ability of an antigen-binding molecule to eliminate antigen from plasma”.
  • the present invention provides methods for increasing the ability of an antigen-binding molecule to eliminate plasma antigen by substituting an amino acid in the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and thereby increasing the FcRn-binding activity at neutral and/or acidic pH as compared to an antigen-binding molecule having an intact FcRn-binding domain.
  • the term “ability to eliminate plasma antigen” means the ability to remove antigen from the plasma when antigen-binding molecules are administered or secreted in vivo.
  • “increase in the ability of antigen-binding molecule to eliminate plasma antigen” herein means that the rate of antigen elimination from the plasma is accelerated upon administration of the antigen-binding molecule as compared to before the modification of the FcRn-binding domain and thus before increasing the human FcRn-binding activity of the antigen-binding molecule in the neutral pH range or before increasing the human FcRn-binding activity and simultaneously reducing its antigen-binding activity in the acidic pH range to less than that in the neutral pH range.
  • the increase in the activity of an antigen-binding molecule to eliminate antigen from the plasma can be assessed, for example, by administering a soluble antigen and an antigen-binding molecule in vivo, and measuring the concentration of the soluble antigen in plasma after administration.
  • concentration of soluble antigen in plasma after administration of the soluble antigen and modified antigen-binding molecule is reduced, the ability of antigen-binding molecule to eliminate plasma antigen can be judged to be increased.
  • a form of soluble antigen can be antigen-binding molecule bound antigen or antigen-binding molecule non-bound antigen whose concentration can be determined as “antigen-binding molecule bound antigen concentration in plasma” and “antigen-binding molecule non-bound antigen concentration in plasma” respectively. The latter is synonymous to “free antigen concentration in plasma”. Since “total antigen concentration in plasma” means the sum of antigen-binding molecule bound antigen and non-bound antigen concentration, or “free antigen concentration in plasma” which is antigen-binding molecule non-bound antigen concentration, the concentration of soluble antigen can be determined as “total antigen concentration in plasma”. Various methods for measuring “total antigen concentration in plasma” or “free antigen concentration in plasma” are well known in the art as described hereinafter.
  • the present invention also provides the use of the FcRn-binding domain of the present invention for improving the pharmacokinetics of antigen-binding molecules. More specifically, the present invention provides methods for improving the pharmacokinetics of the antigen-binding molecule by substituting an amino acid in the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and thereby increasing the FcRn-binding activity at neutral and/or acidic pH as compared to an antigen-binding molecule having an intact FcRn-binding domain.
  • the terms “enhancement of pharmacokinetics”, “improvement of pharmacokinetics”, and “superior pharmacokinetics” can be restated as “enhancement of plasma (blood) retention”, “improvement of plasma (blood) retention”, “superior plasma (blood) retention”, and “prolonged plasma (blood) retention”. These terms are used herein as synonyms.
  • a delayed elimination prolonging the time between administration and elimination of the antigen-binding molecules from plasma as compared to a Control Antigen-binding Molecule (e.g. antigen-binding molecules having an intact FcRn-binding domain); and/or (2) prolonging the plasma retention time of the antigen-binding molecules, preferably in a form in which the antibody or antibody derivative can bind to its antigen after administration of the Antigen-binding molecules as compared to the plasma retention time of a Control Antigen-binding Molecule (e.g.
  • antigen-binding molecules having an intact FcRn-binding domain having an intact FcRn-binding domain
  • Control Antigen-binding Molecule e.g. antigen-binding molecules having an intact FcRn-binding domain
  • a Control Antigen-binding Molecule e.g. antigen-binding molecules having an intact FcRn-binding domain
  • the present invention also provides a method for delaying the elimination of an antigen-binding molecule in a subject, comprising the step of introducing a modification into a FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • improved pharmacokinetics refers not only to prolongation of the period between administration of the antigen-binding molecule to a subject (humans, or non-human animals such as mice, rats, monkeys, rabbits, and dogs) and elimination from the plasma (for example, until the antigen-binding molecule is degraded intracellularly or the like and cannot return to the plasma) to, but also to the prolongation of the plasma retention of the antigen-binding molecule in a form that allows antigen binding (for example, in an antigen-free form of the antigen-binding molecule) during the period from administration until degradation of the antigen-binding molecule.
  • the present invention also provides a method of prolonging the plasma retention time of an antigen-binding molecule, comprising the step of introducing a modification into a FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • Intact human IgG can bind to FcRn from non-human animals.
  • mice administration to mice is preferably used to confirm the property of the antigen-binding molecule of the invention since intact human IgG can bind to mouse FcRn stronger than to human FcRn (Int Immunol. 2001 December; 13(12): 1551-9).
  • mouse in which its native FcRn genes are disrupted and a transgene for human FcRn gene is harbored to be expressed can also be preferably used to be administered in order to confirm the property of the antigen-binding molecule of the invention described hereinafter.
  • “improvement of pharmacokinetics” also includes prolongation of the period between administration and degradation of the antigen-binding molecule during which it is not bound to an antigen (the antigen-free form of antigen-binding molecule).
  • the antigen-binding molecule in plasma cannot bind to a new antigen when the antigen-binding molecule has already bound to an antigen.
  • the longer the period during which the antigen-binding molecule is not bound to an antigen the longer is the period during which it has the potential to bind to a new antigen (the higher the chance of binding to another antigen). In other words, more antigens are bound during a shorter period of time.
  • the plasma concentration of the antigen-free form of antigen-binding molecule can be increased and the total period during which antigen is bound to the antigen-binding molecule can be prolonged by accelerating the antigen elimination from the plasma by administration of the modified antigen-binding molecule.
  • improvement of the pharmacokinetics of antigen-binding molecule includes the improvement of a pharmacokinetic parameter of the antigen-free form of the antigen-binding molecule (any of prolongation of the half-life in plasma, prolongation of mean retention time in plasma, and impairment of plasma clearance), prolongation of the period during which the antigen is bound to the antigen-binding molecule after administration of the modified antigen-binding molecule, and acceleration of antigen-binding molecule-mediated antigen elimination from the plasma.
  • the improvement of pharmacokinetics of antigen-binding molecule can be assessed by determining any one of the parameters, half-life in plasma, mean plasma retention time, and plasma clearance for the antigen-binding molecule or the antigen-free form thereof (“Pharmacokinetics: Enshu ni yoru Rikai (Understanding through practice)” Nanzando).
  • the plasma concentration of the antigen-binding molecule or antigen-free form thereof is determined after administration of the antigen-binding molecule to mice, rats, monkeys, rabbits, dogs, or humans. Then, each parameter is determined.
  • the plasma half-life or mean plasma retention time is prolonged, the pharmacokinetics of the antigen-binding molecule can be judged to be improved.
  • the parameters can be determined by methods known to those skilled in the art.
  • the parameters can be appropriately assessed, for example, by non-compartmental analysis using the pharmacokinetics analysis software WinNonlin (Pharsight) according to the appended instruction manual.
  • the plasma concentration of antigen-free antigen-binding molecule can be determined by methods known to those skilled in the art, for example, using the assay method described in Clin Pharmacol. 2008 April; 48(4): 406-17.
  • the term “improvement of pharmacokinetics” also includes prolongation of the period that an antigen is bound to an antigen-binding molecule after administration of the antigen-binding molecule. Whether the period that antigen is bound to the antigen-binding molecule after administration of the antigen-binding molecule is prolonged can be assessed by determining the plasma concentration of free antigen. The prolongation can be judged based on the determined plasma concentration of free antigen or the time period required for an increase in the ratio of free antigen concentration to the total antigen concentration.
  • the present invention also provides the use of the antigen-binding molecules of the present invention for reducing total or free antigen plasma concentration of a specific antigen, wherein the antigen-binding molecule comprises an antigen-binding domain which can bind said antigen. More specifically, the present invention provides methods for reducing total or free antigen plasma concentration, said method comprising the steps of:
  • an antigen-binding molecule comprising a parent FcRn-binding domain, wherein the antigen-binding molecule comprises an antigen-binding domain which can bind said antigen
  • the present invention provides, comprising the step of introducing a modification into an FcRn-binding domain of said antigen-binding molecule at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • the term “antigen-elimination rate” as used herein refers to the number of antigens that an antigen-binding molecule can remove from the plasma in the time between administration and elimination (i.e. degradation) of the antibody or antibody derivative.
  • the plasma concentration of free antigen not bound to the antigen-binding molecule or the ratio of free antigen concentration to the total concentration can be determined by methods known to those skilled in the art, for example, by the method described in Pharm Res. 2006 January; 23 (1): 95-103.
  • an antigen exhibits a particular function in vivo
  • whether the antigen is bound to an antigen-binding molecule that neutralizes the antigen function can be assessed by testing whether the antigen function is neutralized. Whether the antigen function is neutralized can be assessed by assaying an in vivo marker that reflects the antigen function.
  • Whether the antigen is bound to an antigen-binding molecule that activates the antigen function (agonistic molecule) can be assessed by assaying an in vivo marker that reflects the antigen function.
  • Determination of the plasma concentration of free antigen and ratio of the amount of free antigen in plasma to the amount of total antigen in plasma, in vivo marker assay, and such measurements are not particularly limited; however, the assays are preferably carried out after a certain period of time has passed after administration of the antigen-binding molecule.
  • the period after administration of the antigen-binding molecule is not particularly limited; those skilled in the art can determine the appropriate period depending on the properties and the like of the administered antigen-binding molecule.
  • Such periods include, for example, one day after administration of the antigen-binding molecule, three days after administration of the antigen-binding molecule, seven days after administration of the antigen-binding molecule, 14 days after administration of the antigen-binding molecule, and 28 days after administration of the antigen-binding molecule.
  • plasma antigen concentration means either “total antigen concentration in plasma” which is the sum of antigen-binding molecule bound antigen and non-bound antigen concentration or “free antigen concentration in plasma” which is antigen-binding molecule non-bound antigen concentration.
  • Total antigen concentration in plasma can be lowered by administration of antigen-binding molecule of the present invention by 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, 1,000-fold, or even higher compared to the administration of a reference antigen-binding molecule comprising the intact human IgG Fc region as a human FcRn-binding domain or compared to when antigen-binding domain molecule of the present invention is not administered.
  • Molar antigen/antigen-binding molecule ratio can be calculated as shown below;
  • Molar antigen/antigen-binding molecule ratio can be lowered by administration of antigen-binding molecule of present invention by 2-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 200-fold, 500-fold, 1,000-fold, or even higher as compared to the administration of a reference antigen-binding molecule comprising the intact human IgG Fc region as a human FcRn-binding domain.
  • an intact human IgG1, IgG2, IgG3 or IgG4 is preferably used as the intact human IgG for a purpose of a reference intact human IgG to be compared with the antigen-binding molecules for their human FcRn binding activity or in vivo activity.
  • a reference antigen-binding molecule comprising the same antigen-binding domain as an antigen-binding molecule of the interest and intact human IgG Fc region as a human FcRn-binding domain can be appropriately used.
  • an intact human IgG1 is used for a purpose of a reference intact human IgG to be compared with the antigen-binding molecules for their human FcRn binding activity or in vivo activity.
  • Reduction of total antigen concentration in plasma or molar antigen/antibody ratio can be assessed as described in Examples 6, 8, and 13 of WO2011/122011. More specifically, using human FcRn transgenic mouse line 32 or line 276 (Jackson Laboratories, Methods Mol Biol. (2010) 602: 93-104.), they can be assessed by either antigen-antibody co-injection model or steady-state antigen infusion model when the antigen-binding molecule of interest does not cross-react with the mouse counterpart antigen. When antigen-binding molecule cross-react with mouse counterpart, they can be assessed by simply injecting antigen-binding molecule to human FcRn transgenic mouse line 32 or line 276 (Jackson Laboratories).
  • a mixture of antigen-binding molecule and antigen is administered to the mouse.
  • an infusion pump containing an antigen solution is implanted to the mouse to achieve a constant plasma antigen concentration, and then the antigen-binding molecule is injected to the mouse.
  • the same dosage is used for all administered test antigen-binding molecules.
  • Total antigen concentration in plasma, free antigen concentration in plasma and plasma antigen-binding molecule concentration is measured at an appropriate time point using methods known to those skilled in the art.
  • Total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio can be measured at 2, 4, 7, 14, 28, 56, or 84 days after administration to evaluate the long-term effect of the present invention.
  • a long term plasma antigen concentration is determined by measuring total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio at 2, 4, 7, 14, 28, 56, or 84 days after administration of an antigen-binding molecule in order to evaluate the property of the antigen-binding molecule of the present invention.
  • Whether the reduction of plasma antigen concentration or molar antigen/antigen-binding molecule ratio is achieved by antigen-binding molecule described in the present invention can be determined by the evaluation of the reduction at any one or more of the time points described above.
  • Total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio can be measured at 15 min, 1, 2, 4, 8, 12, or 24 hours after administration to evaluate the short-term effect of the present invention.
  • a short term plasma antigen concentration is determined by measuring total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio at 15 min, 1, 2, 4, 8, 12, or 24 hours after administration of an antigen-binding molecule in order to evaluate the property of the antigen-binding molecule of the present invention.
  • those antigen-binding molecules having a long term effect on activity for eliminating antigen in plasma as described in the present invention have human FcRn-binding activity at pH 7.0 and 25 degrees C. within a range of 28-fold to 440-fold stronger than intact human IgG1 or KD within a range of 3.0 micromolar to 0.2 micromolar.
  • the KD is within a range of 700 nanomolar to 0.2 nanomolar, more preferably, the KD is within a range of 500 nanomolar to 3.5 nanomolar, more preferably, within a range of 150 nanomolar to 3.5 nanomolar.
  • a long term plasma antigen concentration is determined by measuring total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio at 2, 4, 7, 14, 28, 56, or 84 days after administration of an antigen-binding molecule in order to evaluate the long term effect of the antigen-binding molecule of the present invention on activity for eliminating antigen in plasma. Whether the reduction of plasma antigen concentration or molar antigen/antigen-binding molecule ratio is achieved by antigen-binding molecule described in the present invention can be determined by the evaluation of the reduction at any one or more of the time points described above.
  • those antigen-binding molecules having a short term effect on for eliminating antigen in plasma as described in the present invention have human FcRn-binding activity at pH 7.0 and at 25 degrees Celsius 440-fold stronger than intact human IgG or KD stronger than 0.2 micromolar, preferably stronger than 700 nanomolar, more preferably stronger than 500 nanomolar, most preferably, stronger than 150 nanomolar.
  • a short term plasma antigen concentration is determined by measuring total or free antigen concentration in plasma and molar antigen/antigen-binding molecule ratio at 15 min, 1, 2, 4, 8, 12, or 24 hours after administration of an antigen-binding molecule in order to evaluate the short term effect of the antigen-binding molecule of the present invention on activity for eliminating antigen in plasma.
  • Route of administration of an antigen-binding molecule of the present invention can be selected from intradermal, intravenous, intravitreal, subcutaneous, intraperitoneal, parenteral and intramuscular injection.
  • the plasma retention in human is difficult to determine, it may be predicted based on the plasma retention in mice (for example, normal mice, human antigen-expressing transgenic mice, human FcRn-expressing transgenic mice) or monkeys (for example, cynomolgus monkeys).
  • the term “reducing the antigen-binding activity of an antigen-binding molecule in the acidic pH range to less than that in the neutral pH range” means that the antigen-binding activity of the antigen-binding molecule at pH 4.0 to pH 6.5 is impaired as compared to its antigen-binding activity at pH 6.7 to pH 10.0.
  • the above phrase means that the antigen-binding activity of an antigen-binding molecule at pH 5.5 to pH 6.5 is impaired as compared to that at pH 7.0 to pH 8.0, more preferably means that its antigen-binding activity at the early endosomal pH is impaired as compared to its antigen-binding activity at the plasma pH in vivo.
  • the antigen-binding activity of an antigen-binding molecule at pH 5.8 to pH 6.0 is impaired as compared to the antigen-binding activity of the antigen-binding molecule at pH 7.4.
  • the term “reducing the antigen-binding activity of an antigen-binding molecule in the neutral pH range to less than that in the acidic pH range” means that the antigen-binding activity of the antigen-binding molecule at pH 6.7 to pH 10.0 is impaired as compared to its antigen-binding activity at pH 4.0 to pH 6.5.
  • the above phrase means that the antigen-binding activity of an antigen-binding molecule at pH 7.0 to pH 8.0 is impaired as compared to that at pH 5.5 to pH 6.5, more preferably means that its antigen-binding activity at the plasma pH in vivo is impaired as compared to its antigen-binding activity at the early endosomal pH.
  • the antigen-binding activity of an antigen-binding molecule at pH 7.4 is impaired as compared to the antigen-binding activity of the antigen-binding molecule at pH 5.8 to pH 6.0.
  • the expression “reducing the antigen-binding activity of an antigen-binding molecule in the acidic pH range to less than that in the neutral pH range” is also expressed as “increasing the antigen-binding activity of an antigen-binding molecule in the neutral pH range to more than that in the acidic pH range”.
  • the ratio of antigen-binding activity of an antigen-binding molecule between acidic and neutral pH ranges can be increased, for example, by reducing its antigen-binding activity in the acidic pH range, increasing its antigen-binding activity in the neutral pH range, or both.
  • the expression “reducing the antigen-binding activity of an antigen-binding molecule in the neutral pH range to less than that in the acidic pH range” is also expressed as “increasing the antigen-binding activity of an antigen-binding molecule in the acidic pH range to more than that in the neutral pH range”.
  • KD pH7.4
  • KD pH 5.8
  • the ratio of antigen-binding activity of an antigen-binding molecule between acidic and neutral pH ranges can be increased, for example, by reducing its antigen-binding activity in the neutral pH range, increasing its antigen-binding activity in the acidic pH range, or both.
  • the term “reducing the antigen-binding activity (binding ability) at low calcium-ion concentrations to less than its antigen-binding activity at high calcium-ion concentration” as used herein refers to decreasing the binding affinity of the antigen-binding domain for the antigen at a low calcium-ion concentration compared with the binding affinity for the antigen of said antigen-binding domain at a high calcium-ion concentration.
  • the low calcium concentration is preferably 0.5 to 10 micromolar, more preferably 0.1 to 30 micromolar of ionized calcium
  • the high calcium concentration is 100 micromolar to 10 mM, more preferably 200 micromolar to 5 mM of ionized calcium.
  • the expression “impairing the antigen-binding activity in the acidic pH range as compared to that in the neutral pH range” is sometimes used instead of “reducing the antigen-binding activity in the acidic pH range to less than that in the neutral pH range”.
  • the human FcRn-binding activity in the acidic pH range means the human FcRn-binding activity at pH 4.0 to pH 6.5, preferably the human FcRn-binding activity at pH 5.5 to pH 6.5, and particularly preferably the human FcRn-binding activity at pH 5.8 to pH 6.0, which is comparable to the in vivo early endosomal pH.
  • the human FcRn-binding activity in the neutral pH range means the human FcRn-binding activity at pH 6.7 to pH 10.0, preferably the human FcRn-binding activity at pH 7.0 to pH 8.0, and particularly preferably the human FcRn-binding activity at pH 7.4, which is comparable to the in vivo plasma pH.
  • the antigen-binding molecule and uses of the present invention are not limited to any particular theory, the relationship between the reduction (impairment) of the antigen-binding ability of antigen-binding molecule in the acidic pH range to less than that in the neutral pH range and/or the increase (enhancement) of the human FcRn-binding activity in the neutral pH range and the increase in the number of antigens to which a single antigen-binding molecule can bind, due to facilitation of uptake of antigen-binding molecules into cells, and the enhancement of antigen elimination from the plasma can be explained as follows.
  • the antigen-binding molecule is an antibody that binds to a membrane antigen
  • the antibody administered into the body binds to the antigen and then is taken up via internalization into endosomes in the cells together with the antigen while the antibody is kept bound to the antigen. Then, the antibody translocates to lysosomes while the antibody is kept bound to the antigen, and the antibody is degraded by the lysosome together with the antigen.
  • the internalization-mediated elimination from the plasma is called antigen-dependent elimination, and such elimination has been reported with numerous antibody molecules (Drug Discov Today. 2006 January; 11(1-2): 81-8).
  • a single molecule of IgG antibody binds to antigens in a divalent manner, the single antibody molecule is internalized while the antibody is kept bound to the two antigen molecules, and degraded in the lysosome. Accordingly, in the case of typical antibodies, one molecule of IgG antibody cannot bind to three or more molecules of antigen. For example, a single IgG antibody molecule having a neutralizing activity cannot neutralize three or more antigen molecules.
  • IgG molecules The relatively prolonged retention (slow elimination) of IgG molecules in the plasma is due to the function of human FcRn which is known as a salvage receptor of IgG molecules.
  • FcRn human FcRn which is known as a salvage receptor of IgG molecules.
  • IgG molecules When taken up into endosomes via pinocytosis, IgG molecules bind to human FcRn expressed in the endosomes under the acidic condition in the endosomes. While IgG molecules that did not bind to human FcRn transfer to lysosomes where they are degraded, IgG molecules that are bound to human FcRn translocate to the cell surface and return again in the plasma by dissociating from human FcRn under the neutral condition in the plasma.
  • the antigen-binding molecule is an antibody that binds to a soluble antigen
  • the antibody administered into the body binds to the antigen and then is taken up into cells while the antibody is kept bound to the antigen.
  • Many antibodies taken up into cells are released to the outside of the cell via FcRn.
  • the antibodies since the antibodies are released to the outside of the cell, with the antibodies kept bound to antigens, the antibodies cannot bind to antigens again.
  • one molecule of IgG antibody cannot bind to three or more antigen molecules.
  • pH-dependent antigen-binding antibodies that strongly bind to an antigen under the neutral conditions in plasma but dissociate from the antigen under acidic conditions in the endosome (i.e., antibodies that bind under neutral conditions but dissociate under acidic conditions) can dissociate from the antigen in the endosome.
  • pH-dependent antigen-binding antibodies can bind to antigens again when they are recycled to the plasma by FcRn after antigen dissociation; thus, each antibody can repeatedly bind to a number of antigens.
  • the antigen bound to the antigen-binding molecule is dissociated in the endosome and not recycled to the plasma. This facilitates the antigen-binding molecule-mediated antigen uptake into cells.
  • the administration of an antigen-binding molecule can enhance the antigen elimination and thereby reduces the plasma antigen concentration.
  • a calcium concentration-dependent antigen-binding antibody which strongly binds to an antigen under a high calcium concentration condition in plasma, and dissociates from the antigen under a low calcium concentration condition in the endosome, can dissociate from the antigen within the endosome.
  • the calcium concentration-dependent antigen-binding antibody can bind to an antigen again when recycled to plasma via FcRn after antigen dissociation. Thus, such a single antibody can repeatedly bind to multiple antigens. Meanwhile, an antigen bound to the antigen-binding molecule is not recycled to plasma because the antigen dissociates in the endosome, and thus, the antigen-binding molecule promotes uptake of the antigen into cells. The administration of the antigen-binding molecule promotes the elimination of an antigen, and this allows a decrease in the antigen concentration in plasma.
  • the antigen-binding molecule-mediated antigen uptake into cells can be further facilitated by conferring the human FcRn-binding activity under neutral conditions (pH 7.4) to an antibody that binds to an antigen in a pH-dependent manner (binds under neutral conditions but dissociates under acidic conditions).
  • neutral conditions pH 7.4
  • an antibody that binds to an antigen in a pH-dependent manner binds under neutral conditions but dissociates under acidic conditions.
  • the administration of an antigen-binding molecule can enhance the antigen elimination and thereby reduces the plasma antigen concentration.
  • both antibody and antigen-antibody complex are taken up into cells by non-specific endocytosis, and then transported to the cell surface by binding to FcRn under acidic conditions in the endosome. The antibody and antigen-antibody complex are recycled to the plasma via dissociation from FcRn under the neutral condition on cell surface.
  • the antigen elimination rate is presumed to be equal to the rate of antigen uptake into cells via non-specific endocytosis.
  • the pH dependency is insufficient, the antigen that did not dissociate in the endosome is also recycled to the plasma.
  • the rate-determining step in the antigen elimination is the uptake into cells by non-specific endocytosis.
  • IgG-type immunoglobulins which are one of antigen-binding molecules, typically have little FcRn-binding ability in the neutral pH range, but those that exhibit FcRn-binding ability in the neutral pH range could bind to FcRn on the cell surface and thus are taken up into cells in an FcRn-dependent manner by binding to cell-surface FcRn.
  • the rate of FcRn-mediated uptake into cells is more rapid than the rate of uptake into cells by non-specific endocytosis.
  • the rate of antigen elimination can be further accelerated by conferring FcRn-binding ability in the neutral pH range.
  • an antigen-binding molecule having FcRn-binding ability in the neutral pH range transports an antigen into cells more rapidly than the typical (intact human) IgG-type immunoglobulin, and then the antigen-binding molecule is dissociated from the antigen in the endosome.
  • the antigen-binding molecule is recycled to the cell surface or plasma, and again binds to another antigen and is taken up into cells via FcRn. The rate of this cycle can be accelerated by improving FcRn-binding ability in the neutral pH range, thereby accelerating the rate of antigen elimination from the plasma.
  • the efficiency can be further improved by reducing the antigen-binding activity of an antigen-binding molecule in the acidic pH range to less than that in the neutral pH range.
  • the number of antigens to which a single antigen-binding molecule can bind is presumed to increase with an increasing number of cycles achieved by a single antigen-binding molecule.
  • the antigen-binding molecule of the present invention comprises an antigen-binding domain and an FcRn-binding domain.
  • the FcRn-binding domain does not affect antigen binding, or in view of the mechanism described above, facilitation of the antigen-binding molecule-mediated antigen uptake into cells can be expected regardless of the type of antigen, and as a result increases the antigen elimination rate by reducing the antigen-binding activity of an antigen-binding molecule in the acidic pH range (binding ability) to less than that in the neutral pH range and/or increasing its FcRn-binding activity at the plasma pH.
  • the antigen-binding molecules of the present invention may also comprise a substitution at position EU256 in addition to a substitution at the mentioned one or more positions.
  • the amino acid at position EU256 is substituted with a glutamic acid.
  • all foregoing methods of use may also comprise a substitution at position EU256 in addition to a substitution at the one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, whereby the amino acid at position EU256 is preferably substituted with a glutamic acid.
  • the FcRn-binding region is an Fc region in more preferably, it is a human Fc region.
  • substitution in the amino acid sequence of the parent FcRn-binding domain for increasing the FcRn-binding activity at neutral or acidic pH are preferably at position EU252 and EU434 and one at one or more positions selected from the group consisting of EU238, EU250, EU254, EU255, EU256, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, and EU436. More preferably, the substitutions are at three or more positions wherein said three or more positions are one of the combinations set forth in Tables 2, and 4 to 7. Even more preferably, the substitutions are one of the combinations set forth in Table 3
  • the foregoing methods of use may additionally comprise a step introducing an amino acid substitution at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440, and thereby decreasing an increased binding activity for a pre-existing ADA.
  • the substitutions are combinations selected from Table 11.
  • the forgoing methods of use may also comprise an additional step of introducing in an amino acid substitution in an FcRn-binding domain at one or more positions selected from the group consisting of EU234, EU235, EU236, EU237, EU238, EU239, EU265, EU266, EU267, EU269, EU270, EU271, EU295, EU296, EU297 EU298, EU300, EU324, EU325, EU327, EU328, EU329, EU331, and EU332 (according to the EU numbering system).
  • the substitutions L235R/S239K are introduced.
  • the substitutions are combinations selected from Table 14.
  • the antigen-binding molecule of all foregoing uses and method of uses may comprise a pH-dependent antigen-binding domain or a calcium ion-dependent antigen-binding domain.
  • the antigen uptake into cells mediated by the antigen-binding molecules of the present invention is further improved by reducing the antigen-binding activity (binding ability) in the acidic pH range of the above-described antigen-binding molecule to less than its antigen-binding activity in the neutral pH range. Also preferred is the further improvement of the antigen uptake into cells by reducing the antigen-binding activity (binding ability) of the antigen-binding molecule of the present invention at low calcium-ion concentration (i.e.
  • the parent antigen-binding molecule already comprises an ionized calcium-concentration dependent antigen-binding domain.
  • the antigen-binding domain is altered by substituting histidine for at least one amino acid or inserting at least one histidine into the antigen-binding domain of the above-described antigen-binding molecule which facilitates antigen uptake into cells.
  • modified antigen-binding molecule that has an increased FcRn-binding activity at neutral or acidic pH can be decreased when these modifications increase the antigen-binding molecule's affinity for a pre-existing ADA, e.g. the rheumatoid factor.
  • Those antigen-binding molecules of the present invention whose affinity for a pre-existing anti-drug antibody is not significantly increased at a neutral pH as compared to a wild type Fc region, in particular those that comprise in the FcRn binding domain an amino acid substitution at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440, are particularly useful as therapeutic antibodies for treating human patients suffering from an autoimmune disease, transplant rejections (graft-vs.-host disease), other inflammatory diseases and allergic diseases.
  • autoimmune disease is an illness that occurs when body tissue is attacked by its own immune system.
  • autoimmune diseases contemplated herein include systemic lupus erythematosus, lupus nephritis, pemphigoid, pemphigus, dermatomyositis, autoimmune hepatitis, Sjogren syndrome, Hashimoto thyroiditis, rheumatoid arthritis, juvenile (type 1) diabetes, polymyositis, scleroderma, Addison disease, Coeliac disease, Guillain-Barre syndrome, dilated cardiomyopathy, mixed connective tissue disease, Wegener's granulomatosis, anti-phospholipid antibody syndrome, vitiligo, pernicious anemia, glomerulonephritis, and pulmonary fibrosis.
  • autoimmune disease is systemic lupus erythematosus or Lupus nephritis.
  • Transplant rejection include graft-versus-host disease is a process in which a transplant recipient's immune system attacks the transplanted organ or tissue.
  • Other inflammatory and allergic diseases include atherosclerosis and hay fever.
  • an increased binding affinity for a pre-existing ADA can reduce the clinical utility and efficacy of a therapeutic antibody.
  • the utility of a therapeutic antibody can be limited by the pre-existing ADAs, since these ADA can influence their efficacy and pharmacokinetics (e.g. degradation rate).
  • this binding can lead to serious side effects.
  • the present invention provides a method for decreasing the binding activity at neutral pH for a pre-existing ADA of antigen-binding domain comprising an Fc region with an increased binding activity for FcRn at neutral or acidic pH and an increased binding activity for a pre-existing ADA at neutral pH.
  • the present invention also provides the use of the antigen-binding molecules of the present invention comprising a modified FcRn-binding domain for decreasing the binding activity for a pre-existing ADA at neutral pH of an antigen-binding molecule that has an increased affinity for FcRn at neutral or acidic pH and an increased binding activity for a pre-existing ADA.
  • the present invention provides a method for decreasing the binding activity at neutral pH for a pre-existing ADA of an Fc region of an antigen-binding molecule that has an increased binding activity for FcRn at neutral or acidic pH, said method comprising
  • the preferred Fc region in step a) is a human Fc region.
  • the Fc region having an increased binding activity for FcRn at neutral or acidic pH ranges and for pre-existing ADA in the neutral pH ranges comprises a substitution of an amino acid at one or more positions selected from the group consisting of: EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436. More preferably, it comprises a substitution at the positions at any one of the combinations of positions selected from Table 2 and 4 to 7. Even more preferably, it comprises any one of the substitutions or combinations of substitutions set forth in any one of Tables 3 and 17 to 20.
  • step b) comprises substituting an amino acid at any one of the positions in Table 8. More preferably, step b) comprises introducing one of the substitutions or combinations selected from Table 11.
  • the antigen-binding molecule comprises additionally a pH-dependent antigen-binding domain or a calcium ion-dependent antigen-binding domain.
  • the present invention provides a method for decreasing the binding activity for a pre-existing ADA of an antigen-binding molecule that comprises an Fc region having an increased binding activity for FcRn at neutral pH, said method comprising the steps of:
  • the preferred Fc region in step a) is a human Fc region.
  • the Fc region having an increased binding activity for FcRn at neutral pH ranges and for pre-existing ADA in the neutral pH ranges comprises a substitution of an amino acid at one or more positions selected from the group consisting of: EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436.
  • It may also comprise a substitution at position EU256 in addition to a substitution at the one or more positions mentioned above, whereby the amino acid at position EU256 is preferably substituted with a glutamic acid. More preferably, it comprises a substitution at the positions of any one of the position combinations selected from Tables 2 and 4 to 7. Even more preferably, it comprises any one of the substitutions or substitution combinations selected from any one of Table 3 and 17 to 20.
  • step b) comprises substituting an amino acid at any one of the positions in Table 10. More preferably, the positions are selected from the group consisting of a) EU387, b) EU422, c) EU424, d) EU438, e) EU440, f) EU422/EU424, and g) EU438/EU440. Even more preferably, step b) comprises introducing one of the substitutions or combinations selected from Table 11.
  • the present invention provides a method for decreasing the binding activity for a pre-existing ADA of an antigen-binding molecule that comprises an Fc region having an increased binding activity for FcRn at acidic pH, said method comprising the steps of:
  • the preferred Fc region in step a) is a human Fc region.
  • the Fc region having an increased binding activity for FcRn at acidic pH and for pre-existing ADA in the neutral pH ranges comprises an amino acid substitution comprise a substitution
  • the Fc region comprises i) the substitution M434H; or ii) one of the combinations of the group consisting of a) M252Y/S254T/T256E; b) M428L/N434S; and c) T250Q and M428L (EU numbering).
  • step b) an amino acid is substituted at a) position EU424 or b) the positions EU438/EU440. More preferably, the substitutions are a) EU424N or b) the combination EU438R/EU440E.
  • the methods for decreasing the binding activity for a pre-existing ADA further comprises the step c) confirming that said antigen-binding molecule with a modified Fc domain has a decreased binding activity for an endogenous ADA as compared the binding activity for the original antigen-binding molecule as set forth in step a) comprised of an intact Fc domain.
  • the antigen-binding molecule comprises additionally a pH-dependent antigen-binding domain or a Calcium ion-dependent antigen-binding domain.
  • the present invention provides the use of an antigen-binding molecule of the present invention for increasing antigen removal from blood of a mammal, preferably a human patient suffering from an autoimmune disease.
  • the present invention further provides a method for increasing the total number of antigens to which a single antigen-binding molecule can bind without significantly increasing the binding activity for a pre-existing ADA at neutral pH as compared to a parent antibody, said method comprising the steps of
  • step b) altering the parent FcRn binding domain of step a) by substituting an amino acid in the amino acid sequence of the parent FcRn binding domain at one or more of the positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the present invention further provides a method for facilitating the extracellular release of an antigen-free antigen-binding molecule taken up into cells in an antigen-bound form without significantly increasing the binding activity of said antigen-binding molecule for a pre-existing ADA at neutral pH as compared to a parent antibody, comprising the steps of
  • step b) altering the parent FcRn binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, and EU428; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the present invention further provides a method for increasing the ability of an antigen-binding molecule to eliminate plasma antigen without significantly increasing the binding activity for pre-existing ADA at neutral pH compared to parent antibody, said method comprising the steps of
  • step b) altering the parent FcRn binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, and EU428; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the present invention further provides a method for improving the pharmacokinetics of an antigen-binding molecule without significantly increasing the binding activity for a pre-existing ADA at neutral pH as compared to a parent antibody, said method comprising the steps of
  • step b) altering the parent FcRn-binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the present invention further provides a method for reducing total or free antigen plasma concentration without significantly increasing the binding activity for a pre-existing ADA at neutral pH as compared to a parent antibody, said method comprising the steps of
  • an antigen-binding molecule comprising a parent FcRn-binding domain, wherein the antigen-binding molecule comprises an antigen-binding domain which can bind said antigen
  • step b) altering the parent FcRn-binding domain by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU256, EU258 EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436; and c) altering the modified FcRn-binding domain of step b) by substituting an amino acid in the amino acid sequence of the parent FcRn-binding domain at one or more positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440.
  • the preferred Fc region in step a) in the foregoing methods of use is a human Fc region.
  • the amino acid substitution at one or more positions in step b) is a substitution at one or more positions selected from the group consisting of: EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436, whereby the Fc region of step b) has an increased binding activity for FcRn and a pre-existing ADA in the neutral pH ranges.
  • It may also comprise a substitution at position EU256 in addition to a substitution at the one or more positions mentioned above, whereby the amino acid at position EU256 is preferably substituted with a glutamic acid. More preferably, it comprises a substitution at the positions of any one of the position combinations selected from Tables 2 and 4 to 7. Even more preferably, it comprises any one of the substitutions or substitution combinations selected from any one of Table 3 and 17 to 20.
  • step c) comprises substituting an amino acid at any one of the positions in Table 10. More preferably, the positions are selected from the group consisting of a) EU387, b) EU422, c) EU424, d) EU438, e) EU440, f) EU422/EU424, and g) EU438/EU440. Even more preferably, step c) comprises introducing one of the substitutions or combinations selected from Table 11.
  • the amino acid substitution at one or more positions in step b) is a substitution
  • the Fc region of step b) has an increased FcRn-binding activity in the acidic ranges and an increased binding activity for a pre-existing ADA in the neutral pH ranges.
  • the Fc region comprises i) the substitution M434H; or ii) one of the combinations of the group consisting of a) M252Y/S254T/T256E; b) M428L/N434S; and c) T250Q and M428L (EU numbering).
  • step c) an amino acid is substituted at a) position EU424 or b) the positions EU438/EU440. More preferably, the substitutions are a) EU424N or b) the combination EU438R/EU440E.
  • the present invention also relates to pharmaceutical compositions that include antigen-binding molecules of the present invention, or antigen-binding molecules produced by the production methods of the present invention.
  • the antigen-binding molecules of the present invention and antigen-binding molecules produced by the production methods of the present invention have greater activity to reduce plasma antigen concentration by administration as compared to typical antigen-binding molecules, and are therefore useful as pharmaceutical compositions.
  • the pharmaceutical composition of the present invention may include pharmaceutically acceptable carriers.
  • pharmaceutical compositions ordinarily refer to agents for treating or preventing, or testing and diagnosing diseases.
  • compositions of the present invention can be formulated by methods known to those skilled in the art. For example, they can be used parenterally, in the form of injections of sterile solutions or suspensions including water or other pharmaceutically acceptable liquid.
  • such compositions may be formulated by mixing in the form of unit dose required in the generally approved medicine manufacturing practice by appropriately combining with pharmaceutically acceptable carriers or media, specifically with sterile water, physiological saline, vegetable oil, emulsifier, suspension, surfactant, stabilizer, flavoring agent, excipient, vehicle, preservative, binder, or such.
  • the amount of active ingredient may be readily and routinely adjusted to obtain an appropriate amount in a pre-determined range.
  • Sterile compositions for injection can be formulated using vehicles such as distilled water for injection, according to standard formulation practice.
  • Aqueous solutions for injection include, for example, physiological saline and isotonic solutions containing dextrose or other adjuvants (for example, D-sorbitol, D-mannose, D-mannitol, and sodium chloride).
  • dextrose or other adjuvants for example, D-sorbitol, D-mannose, D-mannitol, and sodium chloride.
  • solubilizers for example, alcohols (ethanol and such), polyalcohols (propylene glycol, polyethylene glycol, and such), non-ionic surfactants (polysorbate 80TM, HCO-50, and such).
  • Oils include sesame oil and soybean oils.
  • Benzyl benzoate and/or benzyl alcohol can be used in combination as solubilizers. It is also possible to combine buffers (for example, phosphate buffer and sodium acetate buffer), soothing agents (for example, procaine hydrochloride), stabilizers (for example, benzyl alcohol and phenol), and/or antioxidants. Appropriate ampules are filled with the prepared injections.
  • compositions of the present invention are preferably administered parenterally.
  • the compositions may be in the dosage form for injections, transnasal administration, transpulmonary administration, or transdermal administration.
  • Such compositions may be administered systemically or locally by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, or such.
  • Administration methods can be appropriately selected in consideration of the patient's age and symptoms.
  • the dose of a pharmaceutical composition containing an antigen-binding molecule may be, for example, from 0.0001 to 1,000 mg/kg for each administration. Alternatively, the dose may be, for example, from 0.001 to 100,000 mg per patient. However, the present invention is not limited by the numeric values described above.
  • the doses and administration methods vary depending on the patient's weight, age, symptoms, and such. Those skilled in the art can set appropriate doses and administration methods in consideration of the factors described above.
  • Amino acids contained in the amino acid sequences of the present invention may be post-translationally modified.
  • the modification of an N-terminal glutamine into a pyroglutamic acid by pyroglutamylation is well-known to those skilled in the art.
  • post-translationally modified amino acids are included in the amino acid sequences in the present invention.
  • the present invention provides methods for producing antigen-binding molecules of the present invention.
  • the present invention provides a method for producing antigen-binding molecules having an FcRn-binding domain with an increased binding activity for FcRn at neutral pH as compared to an antigen-binding molecule comprising a wild type Fc region.
  • the present invention provides a method for producing an antigen-binding molecule, which comprises the steps of:
  • the selected antigen-binding molecule comprises an antigen-binding domain that has a lower binding activity for the antigen at a pH 5.5-6.5 than at pH 7-8 or has a calcium dependent antigen binding activity.
  • the FcRn-binding domain of step a) is a FcRn-binding domain of the present invention. More preferably, the FcRn-binding domain comprises an amino acid substations at three or more positions, wherein said three or more positions are one of the combinations set forth in Tables 2, and 4 to 7. Even more preferably, the FcRn-binding domain comprises three or more substitutions wherein said three or more substitutions are one of the combinations set forth in Tables 3, 17 to 20.
  • Steps (a) may comprise substituting an amino acid substitution at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 and selecting a FcRn-binding domain that has stronger human FcRn-binding activity in the neutral pH range than KD 3.2 micromolar.
  • Step (b) may comprise selecting an antigen-binding domain and altering at least one amino acid in the antigen-binding domain as described above in order to get a pH-dependent antigen-binding domain, or selecting a calcium-ion dependent antigen-binding domain.
  • Altering an amino acid is preferably substituting histidine for at least one amino acid or inserting at least one histidine.
  • the site where the at least one histidine mutation is introduced is not particularly limited, and thus it may be introduced at any position as long as the histidine mutation reduces the antigen-binding activity in the acidic pH range to less than that in the neutral pH range. Such histidine mutations may be introduced at a single site or two or more sites.
  • Steps a) and b) may be repeated twice or more times. The number of times of repeating steps (a) and (b) is not particularly limited; however, the number is typically ten times or less.
  • a linker operably linking the FcRn-binding domain and the antigen-binding domain prepared in (a) and (b) is not limited to any form.
  • the human FcRn-binding domain and the antigen-binding domain can be linked by either covalent or non-covalent forces.
  • the linker can be a peptide linker or a chemical linker or a binding pair like a combination of biotin and streptavidin. Modification of a polypeptide including the human FcRn-binding domain and the antigen-binding domain is known in the art.
  • the human FcRn-binding domain and the antigen-binding domain of the present invention can be linked by forming a fusion protein between the human FcRn-binding domain and the antigen-binding domain.
  • genes encoding the human FcRn-binding domain and the antigen-binding domain can be operationally linked so as to form in frame fusion polypeptide.
  • a linker comprising peptide consisting of several amino acids can be inserted between the human FcRn-binding domain and the antigen-binding domain.
  • Various flexible linkers like the linker whose sequence consists of (GGGGS)n (SEQ ID NO: 11) is known in the art.
  • the present invention further provides a method for producing an antigen-binding molecule comprising an FcRn-binding domain with an increased binding activity for FcRn at neutral or acidic pH without a significantly increased binding activity for a pre-existing ADA at neutral pH compared to an antigen-binding molecule comprising a wild type Fc region.
  • the methods for producing an antigen-binding molecule comprising a Fc region with an increased binding activity for FcRn at neutral or acidic pH and a decreased binding activity for an pre-existing ADA at neutral pH, comprises the steps of:
  • the preferred Fc region in step a) is a human Fc region.
  • the Fc region having an increased binding activity for FcRn and pre-existing ADA at neutral or acidic pH ranges and for pre-existing ADA in the neutral pH ranges comprises a substitution of an amino acid at one or more positions selected from the group consisting of: EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436. More preferably, it comprises substitutions at the positions of any one of the position combinations selected from Tables 2 and 4 to 7.
  • step a) includes providing a nucleotide sequence encoding an Fc region having an increased binding activity for FcRn and pre-existing ADA at neutral or acidic pH ranges.
  • the substitutions in step b) amino acid substitutions at one or more positions or a position combination as set forth in Table 10. More preferably, the substitutions of step b) are one of the substitution combinations set forth in Table 11.
  • the amino acid at one or more of the positions selected from the group consisting of EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440 in step b) is preferably substituted by replacing one or more nucleotides in the nucleotide sequence.
  • Steps (b) and (c) may be carried out in either order. Furthermore, step c) may comprise altering at least one amino acid in the antigen-binding domain as described above in order to get a pH-dependent antigen-binding domain, or selecting a calcium-ion dependent antigen-binding domain.
  • altering an amino acid is preferably substituting histidine for at least one amino acid or inserting at least one histidine.
  • the site where the at least one histidine mutation is introduced is not particularly limited, and thus it may be introduced at any position as long as the histidine mutation reduces the antigen-binding activity in the acidic pH range to less than that in the neutral pH range. Such histidine mutations may be introduced at a single site or two or more sites. Steps b) and c) may be repeated twice or more times. The number of times of repeating steps (b) and (c) is not particularly limited; however, the number is typically ten times or less.
  • a linker operably linking the FcRn-binding domain and the antigen-binding domain prepared in (b) and (c) is not limited to any form.
  • the human FcRn-binding domain and the antigen-binding domain can be linked by either covalent or non-covalent forces.
  • the linker can be a peptide linker or a chemical linker or a binding pair like a combination of biotin and streptavidin. Modification of a polypeptide including the human FcRn-binding domain and the antigen-binding domain is known in the art.
  • the human FcRn-binding domain and the antigen-binding domain of the present invention can be linked by forming a fusion protein between the human FcRn-binding domain and the antigen-binding domain.
  • genes encoding the human FcRn-binding domain and the antigen-binding domain can be operationally linked so as to form in frame fusion polypeptide.
  • a linker comprising peptide consisting of several amino acids can be inserted between the human FcRn-binding domain and the antigen-binding domain.
  • Various flexible linkers like the linker whose sequence consists of (GGGGS)n (SEQ ID NO: 11) is known in the art.
  • the production methods of the present invention may further comprise the steps of altering the above-described amino acids and substituting or inserting histidine.
  • non-natural amino acids may be used instead of histidine. Therefore, the present invention can also be understood by replacing the above-mentioned histidine with non-natural amino acids.
  • Step a) of the production methods of the present invention may also comprise a substitution at position EU256 in addition to a substitution at the one or more positions mentioned above, whereby the amino acid at position EU256 is preferably substituted with a glutamic acid.
  • the production methods of the present invention may further comprise a step comprising substituting an amino acid in the amino acid sequence of the Fc region at one or more of the positions selected from the group consisting of EU234, EU235, EU236, EU237, EU238, EU239, EU265, EU266, EU267, EU269, EU270, EU271, EU295, EU296, EU297 EU298, EU300, EU324, EU325, EU327, EU328, EU329, EU331, and EU332 (according to the EU numbering system).
  • the substitutions L235R/S239K are introduced.
  • Parent FcRn-binding domains and antigen-binding molecules comprising them that are used in the production methods of the present invention may be prepared by any method.
  • pre-existing antibodies, pre-existing libraries (phage libraries and the like) antibodies and libraries that are prepared from hybridomas obtained by immunizing animals or from B cells of immunized animals
  • antibodies and libraries prepared by introducing random amino acid alterations into the above-described antibodies and libraries antibodies and libraries prepared by introducing histidine or non-natural amino acid mutations into the above-described antibodies and libraries (libraries with high content of histidine or non-natural amino acid, libraries introduced with histidine or non-natural amino acid at specific sites, and the like), and such.
  • antigen-binding activity and human FcRn binding activity of an antigen-binding molecule can be determined by methods known to those skilled in the art. Conditions except for pH can be appropriately determined by those skilled in the art.
  • the antigen and antigen-binding molecule may bind to each other in any state, and the human FcRn and antigen-binding molecule may bind to each other in any state.
  • the state is not particularly limited; for example, the antigen or human FcRn may be contacted with an immobilized antigen-binding molecule to bind the antigen-binding molecule.
  • the antigen-binding molecule may be contacted with an immobilized antigen or human FcRn to bind the antigen-binding molecule.
  • the antigen-binding molecule may be contacted with the antigen or human FcRn in a solution to bind the antigen-binding molecule.
  • the antigen-binding molecules produced by the above-described methods may be any antigen-binding molecule of the present invention; and preferred antigen-binding molecules include, for example, those having an antigen-binding domain which is an ionized calcium-concentration dependent antigen-binding domain or an antigen-binding domain with histidine substitution for amino acid(s) or insertion of at least one histidine, and said antigen-binding molecule further comprising a human FcRn-binding domain, which comprise an amino acid substitution at one or more positions selected from the group consisting of EU238, EU250, EU252, EU254, EU255, EU258, EU286, EU307, EU308, EU309, EU311, EU315, EU428, EU433, EU434, and EU436 (EU numbering).
  • the antigen-binding molecule of the present invention may also comprise a substitution at position EU256 in addition to a substitution at the one or more positions mentioned above.
  • the amino acid at position EU256 is substituted with a glutamic acid.
  • the FcRn-binding domain comprises an amino acid substations at three or more positions, wherein said three or more positions are one of the combinations set forth in Tables 2, and 4 to 7. Even more preferably, the FcRn-binding domain comprises three or more substitutions wherein said three or more substitutions are one of the combinations set forth in Tables 3, 17 to 20.
  • antigen-binding molecules include for example, those having an antigen-binding domain which is an ionized calcium-concentration dependent antigen-binding domain or an antigen-binding domain with histidine substitution for amino acid(s) or insertion of at least one histidine, and said antigen-binding molecule further comprising a human Fc region with substitutions of the amino acid at one or more of the positions selected from the group consisting of: EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440. More preferably, the FcRn-binding domain contains substitutions of an amino acid in the human FcRn-binding domain at three or more positions wherein said three or more positions are one of the combinations set forth in Tables 9 and 10.
  • a more preferred antigen-binding molecule includes those having those having an antigen-binding domain which is an ionized calcium-concentration dependent antigen-binding domain or an antigen-binding domain with histidine substitution for amino acid(s) or insertion of at least one histidine, and said antigen-binding molecule further comprising a human Fc region with substitutions of the amino acid
  • EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440 (EU numbering).
  • the amino acid at position EU256 is substituted with a glutamic acid.
  • the antigen-binding molecules comprise a substitution combination set forth in Tables 11 to 13.
  • An antibody having a desired activity may be selected by screening from a number of antibodies obtained from the antibody libraries or hybridomas described below.
  • an antigen-binding molecule When altering amino acids in an antigen-binding molecule, it is possible to use a known sequence for the amino acid sequence of an antigen-binding molecule before alteration or the amino acid sequence of an antigen-binding molecule newly identified by methods known to those skilled in the art.
  • the antigen-binding molecule when the antigen-binding molecule is an antibody, it can be obtained from antibody libraries or by cloning an antibody-encoding gene from monoclonal antibody-producing hybridomas.
  • antibody libraries many antibody libraries are already known, and methods for producing antibody libraries are also known; therefore, those skilled in the art can appropriately obtain antibody libraries.
  • phage libraries one can refer to the literature such as Clackson et al., Nature (1991) 352: 624-8; Marks et al., J. Mol. Biol. (1991) 222: 581-97; Waterhouses et al., Nucleic Acids Res. (1993) 21: 2265-6; Griffiths et al., EMBO J. (1994) 13: 324.0-60; Vaughan et al., Nature Biotechnology (1996) 14: 309-14; and Japanese Patent Kohyo Publication No.
  • variable regions of human antibodies can be expressed on the surface of phages as single chain antibodies (scFvs) using phage display methods, and phages that bind to antigens can be selected. Genetic analysis of the selected phages can determine the DNA sequences encoding the variable regions of human antibodies that bind to the antigens.
  • suitable expression vectors can be produced based on these sequences to obtain human antibodies. These methods are already well known, and one can refer to WO 92/01047, WO 92/20791, WO 93/06213, WO 93/11236, WO 93/19172, WO 95/01438, and WO 95/15388.
  • known technologies may be basically used, which involve the use of desired antigens or cells expressing the desired antigens as sensitizing antigens, using these to perform immunizations according to conventional immunization methods, fusing the resulting immune cells with known parent cells by conventional cell fusion methods, screening monoclonal antibody producing cells (hybridomas) by conventional screening methods, synthesizing cDNAs of antibody variable regions (V regions) from mRNAs of the obtained hybridomas using reverse transcriptase, and linking them with DNAs encoding the desired antibody constant regions (C regions).
  • V regions antibody variable regions
  • C regions desired antibody constant regions
  • sensitizing antigens to obtain the above-described antigen-binding molecule genes encoding the H chains and L chains may include, for example, both complete antigens with immunogenicity and incomplete antigens including haptens and the like with no immunogenicity; however they are not limited to these examples.
  • substances comprising polysaccharides, nucleic acids, lipids, and such can be antigens.
  • the antigens of the antigen-binding molecules of the present invention are not particularly limited.
  • the antigens can be prepared by methods known to those skilled in the art, for example, by baculovirus-based methods (for example, WO 98/46777) and such.
  • Hybridomas can be produced, for example, by the method of Milstein et al. (G. Kohler and C. Milstein, Methods Enzymol. (1981) 73: 3-46) and such.
  • immunization may be performed after linking the antigen with a macromolecule having immunogenicity, such as albumin.
  • antigens may be converted into soluble antigens by linking them with other molecules.
  • transmembrane molecules such as membrane antigens (for example, receptors)
  • portions of the extracellular regions of the membrane antigens can be used as a fragment, or cells expressing transmembrane molecules on their cell surface may be used as immunogens.
  • Antigen-binding molecule-producing cells can be obtained by immunizing animals using appropriate sensitizing antigens described above. Alternatively, antigen-binding molecule-producing cells can be prepared by in vitro immunization of lymphocytes that can produce antigen-binding molecules.
  • Various mammals can be used for immunization; such commonly used animals include rodents, lagomorphas, and primates. Such animals include, for example, rodents such as mice, rats, and hamsters; lagomorphas such as rabbits; and primates including monkeys such as cynomolgus monkeys, rhesus monkeys, baboons, and chimpanzees.
  • transgenic animals carrying human antibody gene repertoires are also known, and human antibodies can be obtained by using these animals (see WO 96/34096; Mendez et al., Nat. Genet. (1997) 15: 146-56).
  • desired human antibodies having binding activity against antigens can be obtained by in vitro sensitization of human lymphocytes with desired antigens or cells expressing the desired antigens, and then fusing the sensitized lymphocytes with human myeloma cells such as U266 (see Japanese Patent Application Kokoku Publication No. (JP-B) H01-59878 (examined, approved Japanese patent application published for opposition)).
  • desired human antibodies can be obtained by immunizing transgenic animals carrying a complete repertoire of human antibody genes, with desired antigens (see WO 93/12227, WO 92/03918, WO 94/02602, WO 96/34096, and WO 96/33735).
  • Animal immunization can be carried out by appropriately diluting and suspending a sensitizing antigen in phosphate buffered saline (PBS), physiological saline, or such, and mixing it with an adjuvant to emulsify, if necessary. This is then intraperitoneally or subcutaneously injected into animals. Then, the sensitizing antigen mixed with Freund's incomplete adjuvant is preferably administered several times every four to 21 days. Antibody production can be confirmed by measuring the titer of the antibody of interest in animal sera using conventional methods.
  • PBS phosphate buffered saline
  • Antigen-binding molecule-producing cells obtained from lymphocytes or animals immunized with a desired antigen can be fused with myeloma cells to generate hybridomas using conventional fusing agents (for example, polyethylene glycol) (Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, (1986) 59-103).
  • fusing agents for example, polyethylene glycol
  • hybridoma cells can be cultured and grown, and the binding specificity of the antigen-binding molecule produced from these hybridomas can be measured using known analysis methods, such as immunoprecipitation, radioimmunoassay (RIA), and enzyme-linked immunosorbent assay (ELISA). Thereafter, if necessary, hybridomas producing antigen-binding molecules of interest whose specificity, affinity, or activity has been determined can be subcloned by methods such as limiting dilution.
  • genes encoding the selected antigen-binding molecules can be cloned from hybridomas or antigen-binding molecule-producing cells (sensitized lymphocytes, and such) using probes that can specifically bind to the antigen-binding molecules (for example, oligonucleotides complementary to sequences encoding the antibody constant regions). It is also possible to clone the genes from mRNA using RT-PCR. Immunoglobulins are classified into five different classes, IgA, IgD, IgE, IgG, and IgM.
  • H chains and L chains used in the present invention to produce antigen-binding molecules are not particularly limited and may originate from antibodies belonging to any of these classes or subclasses; however, IgG is particularly preferred.
  • H-chain-encoding genes and L-chain-encoding genes using genetic engineering technologies.
  • Genetically altered antibodies such as chimeric antibodies and humanized antibodies, which have been artificially altered for the purpose of decreasing heterologous immunogenicity and such against humans, can be appropriately produced for antibodies such as mouse antibodies, rat antibodies, rabbit antibodies, hamster antibodies, sheep antibodies, and camel antibodies.
  • Chimeric antibodies are antibodies including H-chain and L-chain variable regions of nonhuman mammal antibody, such as mouse antibody, and the H-chain and L-chain constant regions of human antibody.
  • Chimeric antibodies can be obtained by ligating a DNA encoding a variable region of a mouse antibody to a DNA encoding a constant region of a human antibody, inserting this into an expression vector, and introducing the vector into a host to produce the antibody.
  • a humanized antibody which is also called a reshaped human antibody, can be synthesized by PCR using several oligonucleotides produced so that they have overlapping portions at the ends of DNA sequences designed to link the complementarity determining regions (CDRs) of an antibody of a nonhuman mammal such as a mouse.
  • the resulting DNA can be ligated to a DNA encoding a human antibody constant region.
  • the ligated DNA can be inserted into an expression vector, and the vector can be introduced into a host to produce the antibody (see EP 239400 and WO 96/02576).
  • Human antibody FRs that are ligated via the CDR are selected when the CDR forms a favorable antigen-binding site. If necessary, amino acids in the framework region of an antibody variable region may be replaced such that the CDR of the reshaped human antibody forms an appropriate antigen-binding site (K. Sato et al., Cancer Res. (1993) 53: 10.01-10.06).
  • antibodies may be altered to improve their biological properties, for example, the binding to the antigen.
  • such alterations can be achieved by methods such as site-directed mutagenesis (see for example, Kunkel (1910.0) Proc. Natl. Acad. Sci. USA 82: 488), PCR mutagenesis, and cassette mutagenesis.
  • mutant antibodies whose biological properties have been improved show amino acid sequence homology and/or similarity of 70% or higher, more preferably 80% or higher, and even more preferably 90% or higher (for example, 95% or higher, 97%, 98%, or 99%), when compared to the amino acid sequence of the original antibody variable region.
  • sequence homology and/or similarity is defined as the ratio of amino acid residues that are homologous (same residue) or similar (amino acid residues classified into the same group based on the general properties of amino acid side chains) to the original antibody residues, after the sequence homology value has been maximized by sequence alignment and gap introduction, if necessary.
  • natural amino acid residues are classified into groups based on the characteristics of their side chains as follows:
  • hydrophobic alanine, isoleucine, valine, methionine, and leucine
  • the present invention provides genes encoding the FcRn-binding domain of the present invention and the antigen-binding molecules of the present invention.
  • the genes encoding the antigen-binding molecules of the present invention may be any genes, and may be DNAs, RNAs, nucleic acid analogs, or the like.
  • the present invention also provides host cells carrying the genes described above.
  • the host cells are not particularly limited and include, for example, E. coli and various animal cells.
  • the host cells may be used, for example, as a production system to produce and express the antibodies of the present invention.
  • In vitro and in vivo production systems are available for polypeptide production systems. Such in vitro production systems include, for example, production systems using eukaryotic cells or prokaryotic cells.
  • Eukaryotic cells that can be used as host cells include, for example, animal cells, plant cells, and fungal cells.
  • Animal cells include: mammalian cells, for example, CHO(Chinese hamster ovary cell line), COS (Monkey kidney cell line), myeloma (Sp2/O, NS0 etc), BHK (baby hamster kidney cell line) Hela, Vero, HEK293 (human embryonic kidney cell line with sheared adenovirus (Ad)5 DNA), PER.C6 cell (human embryonic retinal cell line transformed with the Adenovirus Type 5 (Ad5) E1A and E1B genes) 293, etc (see Current Protocols in Protein Science (May, 2001, Unit 5.9, Table 5.9.1)), amphibian cells such as Xenopus laevis oocytes (Valle et al., Nature (1981) 291: 338-340); and insect cells such as Sf9, Sf21, and Tn5.CHO-DG44, CHO
  • Vectors can be introduced into host cells, for example, by calcium phosphate methods, DEAE-dextran methods, methods using cationic liposome DOTAP (Boehringer-Mannheim), electroporation methods, and lipofection methods.
  • Nicotiana tabacum -derived cells and duckweed are known as a protein production system. Calluses can be cultured from these cells to produce the antigen-binding molecules of the present invention.
  • known protein expression systems are those using yeast cells, for example, cells of genus Saccharomyces (such as Saccharomyces cerevisiae and Saccharomyces pombe ); and cells of filamentous fungi, for example, genus Aspergillus (such as Aspergillus niger ). These cells can be used as a host to produce the antigen-binding molecules of the present invention.
  • Bacterial cells can be used in the prokaryotic production systems. Regarding bacterial cells, production systems using Bacillus subtilis are known in addition to the production systems using E. coli described above. Such systems can be used in producing the antigen-binding molecules of the present invention.
  • Genes obtained by the production methods of the present invention are typically carried by (inserted into) appropriate vectors, and then introduced into host cells.
  • the vectors are not particularly limited as long as they stably retain the inserted nucleic acids.
  • preferred cloning vectors include pBluescript vector (Stratagene); however, various commercially available vectors may be used.
  • expression vectors are particularly useful.
  • the expression vectors are not particularly limited as long as the vectors express the antigen-binding molecules in vitro, in E. coli , in culture cells, or in a body of an organism.
  • pBEST vector Promega
  • pET vector Invitrogen
  • pME18S-FL3 vector GenBank Accession No. AB009864
  • pME18S vector Mol Cell Biol. (1988) 8: 466-472
  • EBNA1 protein may be co-expressed to increase the number of copies of the gene of interest.
  • a vector that includes OriP as a initiation site of replication is used (Biotechnol Bioeng. 2001 Oct. 20; 75(2):197-203, Biotechnol Bioeng. 2005 Sep.
  • DNAs of the present invention can be inserted into the vectors by conventional methods, for example, by ligation using restriction enzyme sites (Current protocols in Molecular Biology, edit. Ausubel et al., (1987) Publish. John Wiley & Sons, Section 11.4-11.11).
  • the above host cells are not particularly limited, and various host cells may be used depending on the purpose.
  • Examples of cells for expressing the antigen-binding molecules include bacterial cells (such as those of Streptococcus, Staphylococcus, E. coli, Streptomyces , and Bacillus subtilis ), eukaryotic cells (such as those of yeast and Aspergillus ), insect cells (such as Drosophila S2 and Spodoptera SF9), animal cells (such as CHO, COS, HeLa, C127, 3T3, BHK, HEK293, and Bowes melanoma cells), and plant cells.
  • bacterial cells such as those of Streptococcus, Staphylococcus, E. coli, Streptomyces , and Bacillus subtilis
  • eukaryotic cells such as those of yeast and Aspergillus
  • insect cells such as Drosophila S2 and Spodoptera SF9
  • animal cells such as CHO, COS,
  • Vectors can be introduced into a host cell by known methods, for example, calcium phosphate precipitation methods, electroporation methods (Current protocols in Molecular Biology edit. Ausubel et al. (1987) Publish. John Wiley & Sons, Section 9.1-9.9), lipofection methods, and microinjection methods.
  • the host cells can be cultured by known methods. For example, when using animal cells as a host, DMEM, MEM, RPMI1640, or IMDM may be used as the culture medium. They may be used with serum supplements such as FBS or fetal calf serum (FCS). The cells may be cultured in serum-free cultures. The preferred pH is about 6 to 8 during the course of culturing. Incubation is carried out typically at 30 to 40 degrees C. for about 15 to 200 hours. Medium is exchanged, aerated, or agitated, as necessary.
  • serum supplements such as FBS or fetal calf serum (FCS).
  • FCS fetal calf serum
  • the cells may be cultured in serum-free cultures.
  • the preferred pH is about 6 to 8 during the course of culturing. Incubation is carried out typically at 30 to 40 degrees C. for about 15 to 200 hours. Medium is exchanged, aerated, or agitated, as necessary.
  • Appropriate secretion signals may be incorporated to polypeptides of interest so that the antigen-binding molecules expressed in the host cell are secreted into the lumen of the endoplasmic reticulum, into the periplasmic space, or into the extracellular environment. These signals may be endogenous to the antigen-binding molecules of interest or may be heterologous signals.
  • production systems using animals or plants may be used as systems for producing polypeptides in vivo.
  • a polynucleotide of interest is introduced into an animal or plant and the polypeptide is produced in the body of the animal or plant, and then collected.
  • the “hosts” of the present invention include such animals and plants.
  • the production system using animals include those using mammals or insects. It is possible to use mammals such as goats, pigs, sheep, mice, and bovines (Vicki Glaser SPECTRUM Biotechnology Applications (1993)). The mammals may be transgenic animals.
  • a polynucleotide encoding an antigen-binding molecule of the present invention is prepared as a fusion gene with a gene encoding a polypeptide specifically produced in milk, such as the goat beta-casein.
  • goat embryos are injected with polynucleotide fragments containing the fusion gene, and then transplanted to female goats.
  • Desired antigen-binding molecules can be obtained from milk produced by the transgenic goats, which are born from the goats that received the embryos, or from their offspring. Hormones may be administered as appropriate to increase the volume of milk containing the antigen-binding molecule produced by the transgenic goats (Ebert et al., Bio/Technology (1994) 12: 699-702).
  • Insects such as silkworms may be used to produce the antigen-binding molecules of the present invention.
  • silkworms When silkworms are used, baculoviruses carrying a polynucleotide encoding an antigen-binding molecule of interest can be used to infect silkworms, and the antigen-binding molecule of interest can be obtained from their body fluids.
  • tobacco when plants are used to produce the antigen-binding molecules of the present invention, for example, tobacco may be used.
  • a polynucleotide encoding an antigen-binding molecule of interest is inserted into a plant expression vector, for example, pMON 530, and then the vector is introduced into bacteria, such as Agrobacterium tumefaciens .
  • bacteria such as Agrobacterium tumefaciens .
  • the bacteria are then allowed to infect tobacco such as Nicotiana tabacum , and the desired antigen-binding molecules can be collected from their leaves (Ma et al., Eur. J. Immunol. (1994) 24: 131-138).
  • the desired antigen-binding molecules can be obtained from the duckweed cells (Cox K M et al., Nat. Biotechnol. 2006 December; 24(12): 1591-1597).
  • antigen-binding molecules may be isolated from the inside or outside (such as the medium and milk) of host cells, and purified as substantially pure and homogenous antigen-binding molecules.
  • the methods for isolating and purifying antigen-binding molecules are not particularly limited, and isolation and purification methods usually used for polypeptide purification can be used.
  • Antigen-binding molecules may be isolated and purified, by appropriately selecting and combining, for example, chromatographic columns, filtration, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, and recrystallization.
  • chromatography techniques include, but are not limited to, affinity chromatography, ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse-phase chromatography, and adsorption chromatography (Strategies for Protein Purification and Characterization: A Laboratory Course Manual. Ed Daniel R. Marshak et al., (1996) Cold Spring Harbor Laboratory Press).
  • Such chromatographic methods can be conducted using liquid phase chromatography such as HPLC and FPLC.
  • Columns used for affinity chromatography include, protein A columns and protein G columns. Columns using protein A include, for example, Hyper D, POROS, and Sepharose F. F. (Pharmacia).
  • an antigen-binding molecule can be modified arbitrarily, and peptides can be partially deleted by allowing an appropriate protein modification enzyme to act before or after purification of the antigen-binding molecule.
  • protein modification enzymes include, for example, trypsin, chymotrypsin, lysyl endopeptidases, protein kinases, and glucosidases.
  • Fc regions of the antigen-binding molecule which interacts with FcRn (Nat Rev Immunol. 2007 September; 7(9):715-25) were engineered to have an improved binding affinity to FcRn at neutral pH in order to enhance the antigen elimination from plasma.
  • the mechanism of antigen elimination from plasma by pH-dependent antigen binding antibody with improved binding affinity to FcRn at neutral pH in comparison to the conventional antibody is shown in FIG. 1A .
  • WO2011/122011 disclose mutations (amino acid substitutions) that improve the binding affinity to FcRn at neutral pH and describes Fc variants F1 to F599 (Table 16) that were generated with the focus on improving the binding affinity to FcRn at neutral pH of antibodies.
  • Fc variants F1 to F599 Table 16
  • stability, purity and immunogenicity should be considered.
  • Antibodies which exhibit poor stability and purity are not suitable as a drug, and poor immunogenicity would hinder their clinical development.
  • the variants (IgG1-F600 to IgG-F1434) each comprising a heavy chain prepared as described above and L(WT)-CK (SEQ ID NO: 2) were expressed and purified by the method known to those skilled in the art described in Reference Example 2 of WO2011/122011.
  • hFcRn binding affinity of new Fc variants prepared in Example 1 (F600-F1434) and previous Fc variants prepared in Example 1 of WO2011/122011 (F1-F599) was evaluated using Biacore T100 (GE Healthcare).
  • human FcRn was prepared as described in Reference Example A2.
  • An appropriate amount of protein L (ACTIGEN) was immobilized onto Sensor chip CM4 (GE Healthcare) by the amino coupling method, and the chip was allowed to capture an antibody of interest. Then, diluted FcRn solutions and running buffer (as a reference solution) were injected to allow human FcRn to interact with the antibody captured on the sensor chip.
  • the running buffer used comprised 50 mmol/l sodium phosphate, 150 mmol/l NaCl, and 0.05% (w/v) Tween20 (pH 7.0).
  • FcRn was diluted using each buffer.
  • the chip was regenerated using 10 mmol/l glycine-HCl (pH 1.5).
  • Assays were carried out exclusively at 25 degrees C.
  • the association rate constant ka (1/Ms) and dissociation rate constant k d (1/s), both of which are kinetic parameters, were calculated based on the sensorgrams obtained in the assays, and KD (M) of each antibody for human FcRn was determined from these values.
  • Each parameter was calculated using Biacore T100 Evaluation Software (GE Healthcare). The binding affinity of all Fc variants is shown in Table 16.
  • DSF differential scanning fluorimetry
  • the fluorescence emission was collected at 555 nm with a fixed excitation wavelength at 470 nm. During the DSF experiment, the temperature was increased from 30 to 99 degrees C. and at 0.4 degrees C. increments with an equilibration time of 6 seconds at each temperature prior to measurement. The data were analyzed using Rotor-Gene Q Series Software (QIAGEN). The temperature of the fluorescence transition is defined as the melting temperature (Tm). Tm values of the Fc variants F1-F1434 are shown in Table 16.
  • High molecular weight species percentage (HMW (%)) of the new Fc variants prepared in Example 1 (F600-F1434) and previous Fc variants prepared in Example 1 of WO2011/122011 (F1-F599) was evaluated using size exclusion chromatography (SEC).
  • SEC was performed in ACQUITY UPLC H-Class system (waters). The antibodies were injected onto a BEH200 SEC column (1.7 micrometer, 4.6 ⁇ 150 mm, waters). The mobile phase was 0.05 M sodium phosphate, 0.3 M sodium chloride (pH7.0, Isekyu), running isocratically at a flow rate of 0.3 mL/min. Eluted protein was detected by UV absorbance at 215 nm. The data were analyzed using Empower2 (waters). Peaks eluting earlier than the antibody monomer peak were recorded in the HMW components percentile. The HMW (%) of all Fc variants (F1-F1434) are shown in Table 16.
  • ADAs anti-drug antibodies
  • Clinical utility and efficacy of the therapeutic antibodies can be limited by the production of anti-drug antibodies (ADAs), since ADA can influence their efficacy and pharmacokinetics and sometimes lead to serious side effects.
  • ADAs anti-drug antibodies
  • a number of reports describe the importance of effector T-cell epitopes present in the therapeutic protein.
  • T-cell epitopes such as Epibase (Lonza), iTope/TCED (Antitope) and EpiMatrix (EpiVax) have been developed.
  • Epibase LiTope/TCED (Antitope)
  • EpiMatrix EpiMatrix
  • Epibase Light was used to evaluate the potential immunogenicity of the Fc variants.
  • Epibase Light is an in silico tool to calculate the binding affinity of 9-mer peptide to major DRB1 alleles using FASTER algorism (Expert Opin Biol Ther. 2007 March; 7(3):405-18.).
  • Epibase Light identifies T-cell epitopes with strong binding and medium binding to MHC class II.
  • DRB1 allotype population frequency used in the formula following DRB1 allotype population frequency based on Caucasian population was used.
  • DRB1*0701 25.3%
  • DRB1*1501 (23.1%)
  • DRB1*0301 (21.7%)
  • DRB1*0101 15.3%
  • DRB1*0401 (13.8%)
  • DRB1*1101 11.8%
  • DRB1*1302 8.8%
  • DRB1*1401 4.9%)
  • DRB1*0901 1.
  • the total number of any strong and medium binding epitopes identified in constant region (CH1-hinge-CH2-CH3) of the variants by FASTER algorism was used as number of critical epitopes in the formula.
  • Filtered epitopes are those with human antibody germline sequence or junction regions between variable region and constant region, and only non-filtered epitopes are considered (counted as critical epitope) in the immunogenicity score calculation.
  • Immunogenicity score of amino acid sequence of new Fc variants described in Example 1 was calculated using above described Epibase Light (Lonza) system. Immunogenicity score of all Fc variants (F1-F1434) are shown in Table 16.
  • hFcRn binding affinity and Tm of previous Fc variants (F1-F599) described in Example 1 of WO2011/122011 and new Fc variants (F600-F1052) generated and evaluated in Example 1 were plotted and are shown in FIG. 2 .
  • hFcRn binding affinity and HMW (%) of previous and new Fc variants were plotted and are shown in FIG. 3 .
  • hFcRn binding affinity and immunogenicity score of Fc variants F1-F599 and new Fc variants (F600-F1052) were plotted and are shown in FIG. 4 .
  • the new Fc variants (F600-F1052) and previous Fc variants (F1-F599) variants having Ser239Lys or Asp270Phe mutation were deleted from the plots. Since Ser239Lys and Asp270Phe mutation improved the stability (Tm) while it did not improve FcRn binding affinity and reduced the binding affinity to all human Fc gamma receptors, in the following detailed analysis of Group 1-4, stability of Fc variants should be compared within the variants that do not have Ser239Lys nor Asp270Phe mutation.
  • new Fc variants F600-F1052 and previous Fc variants (F1-F599) variants having Pro257Xxx (Xxx is Ala or Val or Ile or Leu or Thr) or Met252Trp mutation were deleted from the plots although these variants improve FcRn binding affinity.
  • Pro257Xxx and Met252Trp mutation did not exhibit significant reduction in Tm suggesting that variants with Pro257Xxx and Met252Trp mutation have high stability. Nevertheless, these variants having Pro257Xxx or Met252Trp mutations showed significant aggregation and precipitation during an accelerated stability study or when stored refrigerated. Due to their detrimental stability, Fc variants with Pro257Xxx and Met252Trp mutation are not acceptable for pharmaceutical development and therefore, in the following detailed analysis of Group 1-4, such Fc variants should be deleted from the plots.
  • New Fc variants (F600-F1052) generated and evaluated in Example 1, and previous Fc variants (F1-F599) described in Example 1 of WO2011/122011, with binding affinity to hFcRn stronger than 15 nM (described as Group 1 hereafter), were analyzed in detail by plotting hFcRn binding affinity in X-axis and Tm, HMW (%) and immunogenicity score in Y-axis.
  • Tm criteria was set as higher than 57.5 degrees C.
  • HMW (%) criteria was set as lower than 2%
  • immunogenicity score was set as lower than 500.
  • Fc variants in Group 1 which satisfies all the developability criteria (Tm higher than 57.5 degrees C., HMW (%) lower than 2%, and immunogenicity score lower than 500) are shown in Table 17.
  • New Fc variants (F600-F1052) generated and evaluated in Example 1, and previous Fc variants (F1-F599) described in Example 1 of WO2011/122011, with binding affinity to hFcRn between 15 nM and 50 nM (hereafter called “Group 2”), were analyzed in detail by plotting hFcRn binding affinity on the X-axis and Tm, HMW (%) and immunogenicity score on Y-axis.
  • Tm criteria was set as higher than 60 degrees C.
  • HMW (%) criteria was set as lower than 2%
  • immunogenicity score was set as lower than 500.
  • Fc variants in Group 2 which satisfies all the developability criteria (Tm higher than 60 degrees C., HMW (%) lower than 2%, and immunogenicity score lower than 500) are shown in Table 18.
  • New Fc variants (F600-F1052) generated and evaluated in Example 1, and previous Fc variants (F1-F599) described in Example 1 of WO2011/122011, with binding affinity to hFcRn between 50 nM and 150 nM (called hereinafter “Group 3”), were analyzed in detail by plotting hFcRn binding affinity on the X-axis and Tm, HMW (%) and immunogenicity score on the Y-axis.
  • Tm criteria was set as higher than 63.0 degrees C.
  • HMW (%) criteria was set as lower than 2%
  • immunogenicity score was set as lower than 250.
  • Fc variants in Group 3 which satisfies all the developability criteria (Tm higher than 63.0 degrees C., HMW (%) lower than 2%, and immunogenicity score lower than 250) are shown in Table 19.
  • New Fc variants (F600-F1052) generated and evaluated in Example 1, and previous Fc variants (F1-F599) described in Example 1 of WO2011/122011, with binding affinity to hFcRn between 150 nM and 700 nM (called hereinafter “Group 4”), were analyzed in detail by plotting hFcRn binding affinity on X-axis and Tm, HMW (%) and immunogenicity score on Y-axis.
  • Tm criteria was set as higher than 66.5 degrees C.
  • HMW (%) criteria was set as lower than 2%
  • immunogenicity score was set as lower than 250.
  • Fc variants in Group 4 which satisfies all the developability criteria (Tm higher than 66.5 degrees C., HMW (%) lower than 2%, and immunogenicity score lower than 250) are shown in Table 20.
  • new Fc variants described in Table 17 to 20 have high Tm, low HMW (%), and low immunogenicity score which are suitable for pharmaceutical development of antigen-binding molecule capable of removing antigen from the plasma.
  • Fv-4-IgG1 comprising VH3-IgG1 (SEQ ID NO: 1) and VL3-CK (SEQ ID NO: 3)
  • previous Fc variant Fv-4-F11 comprising VH3-F11 (SEQ ID NO: 4) and VL3-CK (SEQ ID NO: 3)
  • new Fc variants Fv-4-F652 comprising VH3-F652 (SEQ ID NO: 5) and VL3-CK (SEQ ID NO: 3)
  • Fv-4-F890 comprising VH3-F890 (SEQ ID NO: 6) and VL3-CK (SEQ ID NO: 3
  • Fv-4-F946 comprising VH3-F946 (SEQ ID NO: 7) and VL3-CK (SEQ ID NO: 3) were expressed and purified by the method known to those skilled in the art described in Reference Example 2 of WO2011/122011.
  • Anti-human IL-6 receptor antibodies were administered to the model animals to assess the in vivo dynamics after administration of soluble human IL-6 receptor.
  • Monoclonal anti-mouse CD4 antibody (in house) was administered at 20 mg/kg before implanting infusion pump, and 7 and 17 days after antibody administration into the caudal vein to suppress the production of neutralizing antibody against soluble human IL-6 receptor.
  • an infusion pump containing 92.8 microgram/ml soluble human IL-6 receptor was implanted under the skin on the back of the mice. Three days after implantation of an infusion pump, anti-human IL-6 receptor antibodies were administered once into the caudal vein.
  • Fv-4-IgG1, Fv-4-F652, Fv-4-F890 and Fv-4-F946 were administered at as dosage of 1 mg/kg together with approximately 1 g/kg Sanglopor (CSL Behring), and in study 2, Fv-4-IgG1, Fv-4-F11 and Fv-4-F652 were administered at 1 mg/kg.
  • no antibody was administered to the control group (no antibody injection).
  • Blood was collected at appropriate time points after the administration of the anti-human IL-6 receptor antibody. The collected blood was immediately centrifuged at 15,000 rpm and 4 degrees C. for 15 minutes to separate plasma. The separated plasma was stored in a refrigerator at ⁇ 20 degrees C. or below before the assay.
  • the concentration of anti-human IL-6 receptor antibody in mouse plasma was measured by ELISA.
  • Anti-human IgG (gamma-chain specific) F(ab′)2 antibody fragment (Sigma) was dispensed onto a Nunc-ImmunoPlate MaxiSorp (Nalge Nunc International) and allowed to stand overnight at 4 degrees C. to prepare anti-human IgG-immobilized plates.
  • Calibration curve samples having plasma concentrations of 0.8, 0.4, 0.2, 0.1, 0.05, 0.025, and 0.0125 microgram/ml, and mouse plasma samples diluted 100-fold or more were prepared.
  • hsIL-6R 200 microliter of 20 ng/ml hsIL-6R were added to 100 microliter of the calibration curve samples and plasma samples, and then the samples were allowed to stand for one hour at room temperature. Subsequently, the samples were dispensed onto the anti-human IgG-immobilized plates, and allowed to stand for one hour at room temperature. Then, Biotinylated Anti-Human IL-6R Antibody (R&D) was added to react for one hour at room temperature.
  • R&D Biotinylated Anti-Human IL-6R Antibody
  • Streptavidin-PolyHRP80 (Stereospecific Detection Technologies) was added to react for one hour at room temperature, and chromogenic reaction was carried out using TMP One Component HRP Microwell Substrate (BioFX Laboratories) as a substrate. After stopping the reaction with 1 N sulfuric acid (Showa Chemical), the absorbance at 450 nm was measured by a microplate reader. The concentration in mouse plasma was calculated from the absorbance of the calibration curve using the analytical software SOFTmax PRO (Molecular Devices).
  • hsIL-6R concentration was measured by electrochemiluminescence.
  • hsIL-6R calibration curve samples adjusted to concentrations of 2,000, 1,000, 500, 250, 125, 62.5, and 31.25 pg/ml, and mouse plasma samples diluted 50-fold or more were prepared.
  • the samples were mixed with a solution of Monoclonal Anti-human IL-6R Antibody (R&D) ruthenium-labeled with Sulfo-Tag NHS Ester (Meso Scale Discovery), Biotinylated Anti-human IL-6R Antibody (R&D, Systems Inc., USA), and tocilizumab (Chugai Pharmaceutical Co., Ltd.)), and then allowed to react overnight at 37 degrees C.
  • R&D Monoclonal Anti-human IL-6R Antibody
  • Sulfo-Tag NHS Ester Sulfo-Tag NHS Ester
  • the final concentration of tocilizumab as an anti-human IL-6 receptor antibody was 333 microgram/ml, which is in excess of the concentration of anti-human IL-6 receptor antibody contained in the samples, for the purpose of binding nearly all of the hsIL-6R molecules in the samples to tocilizumab.
  • the samples were dispensed into an MA400 PR Streptavidin Plate (Meso Scale Discovery), and allowed to react for one hour at room temperature, and washing was performed.
  • Read Buffer T ( ⁇ 4) (Meso Scale Discovery) was dispensed, the measurement was performed by the Sector PR 400 Reader (Meso Scale Discovery).
  • the hsIL-6R concentration was calculated based on the response of the calibration curve using the analytical software SOFTmax PRO (Molecular Devices).
  • FIG. 17 shows plasma hsIL-6R concentration time profile
  • FIG. 18 shows plasma antibody concentration time profile after injection of Fv-4-IgG1, Fv-4-F652, Fv-4-F890 and Fv-4-F946.
  • Fv-4-IgG1 and control no antibody injection
  • Fv-4-F652, Fv-4-F890 and Fv-4-F946 having new Fc variants with improved binding to FcRn at neutral pH exhibited significant reduction of plasma hsIL-6R concentration demonstrating in vivo antigen elimination effect of pH-dependent antigen binding antibody with improved binding to FcRn at neutral pH.
  • Fv-4-F652 and Fv-4-F890 demonstrated 30-fold and 10-fold antigen elimination effect at day 7 compared to Fv-4-IgG1, respectively, plasma antibody concentration time profile of Fv-4-F652 and Fv-4-F890 were comparable to Fv-4-IgG1.
  • Fv-4-F652 and Fv-4-F890 were able to selectively eliminate soluble antigen from plasma while maintaining antibody pharmacokinetics comparable to that of Fv-4-IgG1.
  • Fv-4-F890 belongs to Group 3, and this study demonstrated that Fc variants in Group 3 can reduce then plasma antigen concentration by approximately 10-fold while maintaining the antibody pharmacokinetic comparable to IgG1. This means that applying Group 3 Fc variant to pH-dependent antigen binding IgG1 antibody can lower the antibody dosage by 10-fold. Such reduction in antibody dosage by Group 3 Fc variant is especially meaningful when antibody dosage needs to be reduced, and simultaneously requires infrequent dosing.
  • Fv-4-F946 demonstrated 100-fold reduction of plasma hsIL-6R concentration compared to Fv-4-IgG1, and antibody clearance of Fv-4-F946 was larger than Fv-4-IgG1.
  • Fv-4-F946 belongs to Group 2, and this study demonstrated that Fc variants in Group 2 can reduce the plasma antigen concentration by approximately 100-fold although the antibody clearance is larger than IgG1. This means that applying Group 2 Fc variant to pH-dependent antigen binding IgG1 antibody can reduce the total plasma antigen concentration by approximately 100-fold.
  • target plasma antigen concentration is too high to neutralize by realistic antibody dosage (i.e 100 mg/kg)
  • 100-fold reduction of total antigen concentration regardless of the increase of antigen clearance by Group 2 Fc variant means that target antigen can be neutralized by less than 10 mg/kg, which is a realistic antibody dosage.
  • Previous studies described in Example 1 of WO2011/122011 revealed that the extent of antigen elimination and antibody clearance correlated with binding affinity to FcRn at neutral pH. As shown in FIG. 17 , Fv-4-F652 exhibited larger extent of antigen elimination compared to Fv-4-F890 although antibody pharmacokinetics was comparable to Fv-4-F890. Therefore, it was suggested that specific mutation in F652 contributed to enhanced antigen sweeping effect.
  • FIG. 19 shows plasma hsIL-6R concentration time profile and FIG. 20 shows plasma antibody concentration time profile after injection of Fv-4-IgG1, Fv-4-F11 and Fv-4-F652.
  • Fv-4-F11 exhibited reduction of plasma hsIL-6R concentration
  • Fv-4-F652 exhibited larger reduction of plasma hsIL-6R concentration
  • Fv-4-F11 and Fv-4-F652 exhibited comparable plasma antibody concentration time profile.
  • Pro238Asp mutation were able to enhance antigen elimination plasma while maintaining antibody pharmacokinetics comparable to Fv-4-IgG1. Therefore, Pro238Asp mutation is extremely valuable for enhancing antigen elimination by pH-dependent antigen binding antibody.
  • ADAs anti-drug antibodies
  • presence of pre-existing antibodies against therapeutic antibody can also be problematic from the point of ADA.
  • therapeutic antibody for patients with autoimmune disease such as rheumatoid arthritis, rheumatoid factor, an autoantibody against human IgG, could be an issue of pre-existing antibody.
  • Rheumatoid factor is a polyclonal autoantibody against human IgG, and their epitope in human IgG varies among the clone, but their epitope seems to be located in the CH2/CH3 interface region as well as CH3 domain which could overlap with the FcRn binding epitope. Therefore, mutations to increase the binding affinity to FcRn might also increase the binding affinity to specific clone of rheumatoid factor.
  • M252Y/S254T/T256E (YTE) variant J Biol Chem 2006 281:23514-23524.
  • M428L/N434S (LS) variant Nat Biotechnol, 2010 28:157-159.
  • N434H variant Clinical Pharmacology & Therapeutics (2011) 89(2):283-290.
  • Fc regions of the antigen-binding molecule which interacts with FcRn (Nat Rev Immunol. 2007 September; 7(9):715-25) was engineered to have improved binding affinity to FcRn at neutral pH, such engineered Fc variants include F11 variant, F68 variant, F890 variant and F947 variant.
  • FIG. 1 The mechanism of antigen elimination from plasma by pH-dependent antigen binding antibody with improved binding affinity to FcRn at neutral pH in comparison to the conventional antibody is shown in FIG. 1 .
  • Such Fc variant with improved FcRn-binding could exhibit increased binding to rheumatoid factor as in the case of previously reported Asn434H is mutation. Therefore, we tested whether these FcRn binding improved Fc variants would exhibit increased binding to rheumatoid factor.
  • Variant antibodies used in the following study were Fv-4-hIgG1, Fv-4-YTE, Fv-4-LS, Fv-4-N434H, Fv-4-F11, Fv-4-F68, Fv-4-890 and Fv-4-F947.
  • Binding assay against rheumatoid factor was performed by Electrochemiluminescence (ECL) at pH7.4.
  • the assays were performed with the serum of 15 or 30 individual RA patients (Proteogenex). 50-fold diluted serum samples, Biotin labeled test antibody (1 microgram/mL) and SULFO-TAG NHS Ester (Meso Scale Discovery) labeled test antibody (1 microgram/mL) were mixed and incubated for 3 hr at room temperature. Then, the mixtures were added to Streptavidin coated MULTI-ARRAY 96 well plates (Meso Scale Discovery), and the plates were incubated for 2 hr at room temperature and washed. After Read Buffer T ( ⁇ 4) (Meso Scale Discovery) was added to each well, plates were immediately set on the SECTOR imager 2400 Reader (Meso Scale Discovery) and the chemiluminescence was measured.
  • ECL Electrochemiluminescence
  • FIGS. 21 and 22 are the ECL response of the serum from 15 or 30 individual RA patients.
  • Fv-4-hIgG1 with native human IgG1 FIGS. 21-1 and 22-1 ) showed only weak rheumatoid factor binding, whereas all the FcRn binding improved Fc variants (Fv-4-YTE ( FIG. 21-2 ), Fv-4-LS ( FIG. 21-3 ), Fv-4-N434H ( FIG. 22-2 ), Fv-4-F11 ( FIG. 22-3 ), Fv-4-F68 ( FIG. 22-4 ), Fv-4-890 ( FIG. 22-5 ) and Fv-4-F947 ( FIG.
  • FIG. 23 shows the mean, geomean and median of the ECL response of the serum of the above mentioned antibody variants with the blood of fifteen RA patients.
  • Fv-4-F890 was selected as parent Fc variant, and single mutation and combined Fc variants of single mutation were introduced into Fv-4-F890.
  • the novel Fc variants F1058 to F1073, F1107 to F1114, F1104 to F1106, and F1230 to F1232 described in Table 21 were generated.
  • Fv-4-F947 was selected as parent Fc variant and same single and combined mutations were introduced.
  • the novel Fc variants F1119-F1124 described in Table 21 were generated. First the variants were evaluated for their binding affinity to human FcRn at pH7.0. Results are also described in Table 21. Compared to either parent Fv-4-F890 or Fv-4-F947, these variants did not show significant reduction in binding affinity against human FcRn, demonstrating that these mutations did not affect human FcRn binding.
  • FIGS. 24 to 29 show the ECL response of the serum from fifteen individual RA patients for the following variants of the antibody: Fv-4-IgG1, Fv-4-F890, Fv-4-F1058 to Fv-4-1073 ( FIG. 24 ), Fv-4-F1104 to Fv-4-F1106 ( FIG. 26 ), Fv-4-F1107 to Fv-4-F1114 ( FIG. 27 ), Fv-4-F1230 to Fv-4-F1232 ( FIG. 28 ), Fv-4-947 and Fv-4-F1119 to Fv-4-F1124 ( FIG. 29 ).
  • FIGS. 25-1, 25-2 and 25-3 are the mean, geomean and median of the ECL response of the serum from fifteen RA patients for the variants Fv-4-IgG1, Fv-4-F890, and Fv-4-F1058 to Fv-4-1073.
  • novel Fc variants with single mutation to F890 such as F1062, F1064-F1072 and F1107-F1114 exhibited significant reduction in rheumatoid factor binding.
  • F1062, F1064, F1068, F1070, F1072, F1107 to F1109 and F1111-F1113 exhibited comparable rheumatoid factor binding as native IgG1 demonstrating that the increased immunogenicity risk of F890 variant was completely eliminated by introducing additional single mutation to reduce rheumatoid factor binding without affecting human FcRn binding. Since rheumatoid factor in patients is a polyclonal antibody binding to multiple epitopes in the Fc region, it was surprising that a single mutation significantly eliminated the binding of rheumatoid factor to the Fc region.
  • double mutated Fc F1106 (Q438R/S440E) showed significant reduction in rheumatoid factor binding.
  • double mutated Fc F1230 (Q438R/S440D), F1231 (Q438K/S440E) and F1232 (Q438K/S440D) also showed additional reduction in rheumatoid factor binding by combination of mutations.
  • F1104 (V422E/S424R) or F1105 (V422S/S424R) did not show any combination effect.
  • Fv-4-F939 selected as parent Fc variant, other mutations for increasing FcRn binding (S254T or T256E) and for reducing rheumatoid factor binding (H433D) were evaluated.
  • Novel Fc variants F1291, F1268, F1269, F1243, F1245, F1321, F1340 and F1323 described in Table 22 were generated.
  • the variants were evaluated for their binding affinity to human FcRn at pH7.0. Results are also described in Table 22.
  • Q438R/S440E, Q438K/S440D and Q438K/S440E mutations showed significant reduction in rheumatoid factor binding with variants having other mutations for increasing FcRn binding (S254T or T256E).
  • the variants were evaluated for their binding affinity to human FcRn at pH7.0 and the presence of additional glycosylation by SDS-Page. Results are described in Table 23. F1077 (K248N), F1080 (S424N), F1081 (Y436N/Q438T) and F1082 (Q438N) were found to have additional glycosylation, and especially F1080 (S424N) maintained binding affinity against human FcRn.
  • Fv-4-N434H and Fv-4-LS variants which improves FcRn binding at acidic pH and prolongs antibody pharmacokinetics
  • Q438R/S440E mutations or S424N mutation were introduced into these variants.
  • Novel Fc variants F1166, F1167, F1172, F1173, F1170 and F1171 described in Table 25 were generated. First the variants were evaluated for their binding affinity to human FcRn at pH6.0. Results are also described in Table 25.
  • mutation such as Pro387Arg, Val422Glu, Val422Arg, Val422Ser, Val422Asp, Val422Lys, Val422Thr, Val422Gln, Ser424Glu, Ser424Arg, Ser424Lys, Ser424Asn, Ser426Asp, Ser426Ala, Ser426Gln, Ser426Tyr, His433Asp, Tyr436Thr, Gln438Glu, Gln438Arg, Gln438Ser, Gln438Lys, Ser440Glu, Ser440Asp, Ser440Gln (positions are given in EU numbering) are extremely useful for reducing the immunogenicity of antigen-binding molecule containing FcRn binding increased Fc region (for example F1-F1434) such as pH-dependent antigen binding antibody with improved binding affinity to FcRn at neutral pH which is capable of eliminating antigen from plasma and antibody with improved binding affinity to FcRn at acidic pH which
  • Mutation sites other than EU387, EU422, EU424, EU426, EU433, EU436, EU438 and EU440 for reducing the binding of rheumatoid factor without affecting human FcRn binding could be selected from 248-257, 305-314, 342-352, 380-386, 388, 414-421, 423, 425-437, 439, and 441-444 in EU numbering.
  • Novel Fc variants F939, F1378, F1379, F1262, F1138, F1344, F1349, F1350, F1351, F1261, F1263, F1305, F1306, F1268, F1269, F1413, F1416, F1419, F1420, F1370, F1371, F1599, F1600, F1566, F1448, F1601-F1603, F1531, F1604, F1605, F1586, F1592, F1610-F1615, F1567, F1572, F1576, F1578, F1579, F1641-F1655, F1329, F1331) described in Table 27 were generated. First the variants were evaluated for their binding affinity to human FcRn at pH7.0. Results are also described in Table 27.
  • Double mutations for decreasing rheumatoid factor binding showed significant reduction in rheumatoid factor binding to other mutations for increasing FcRn binding at neutral pH.
  • Novel Fc variants (F1718-F1721, F1671, F1670, F1711-F1713, F1722-F1725, F1675, F1714-F1717, F1683, F1756-F1759, F1681, F1749-F1751, F1760-F1763, F1752-F1755, F1685) described in Table 28 were generated. First the variants were evaluated for their binding affinity to human FcRn at pH6.0. Results are also described in Table 28.
  • Double mutations for decreasing rheumatoid factor binding showed significant reduction in rheumatoid factor binding to other mutations for increasing FcRn binding at acidic pH.
  • Fv-4-IgG1 comprising VH3-IgG1 (SEQ ID NO: 1) and VL3-CK (SEQ ID NO: 3)
  • new Fc variants Fv-4-F1243 comprising VH3-F1243 (SEQ ID NO: 8) and VL3-CK (SEQ ID NO: 3)
  • Fv-4-F1245 comprising VH3-F1245 (SEQ ID NO: 9) and VL3-CK (SEQ ID NO: 3) were expressed and purified by the method known to those skilled in the art described in Example 2 of WO2011/122011.
  • Fv-4-F1243 and Fv-4-F1245 have novel Fc region with improved binding affinity to human FcRn at neutral pH, but significantly reduced binding to rheumatoid factor.
  • an in vivo study of Fv-4-IgG1, Fv-4-F1243 and Fv-4-F1245 was performed in a human IL-6 receptor steady-state infusion model using human FcRn transgenic mice.
  • FIG. 131 shows plasma hsIL-6R concentration time profile and FIG. 132 shows plasma antibody concentration time profile after injection of Fv-4-IgG1, Fv-4-F1243 and Fv-4-F1245.
  • Fv-4-IgG1 and control no antibody injection
  • Fv-4-F1243 and Fv-4-F1245 having novel Fc variants with improved binding to FcRn at neutral pH exhibited significant reduction of plasma hsIL-6R concentration demonstrating in vivo antigen elimination of pH-dependent antigen binding antibody with improved binding to FcRn at neutral pH.
  • Fv-4-F1243 and Fv-4-F1245 demonstrated 10-fold antigen elimination effect at day 21 or day 7 compared to Fv-4-IgG1, respectively, whereby the plasma antibody concentration time profile of Fv-4-F1243 and Fv-4-F1245 was comparable to Fv-4-IgG1.
  • pH-dependent anti-human IL6 receptor IgG1 antibody Fv-4-IgG1 comprising VH3-IgG1 (SEQ ID NO: 1) and VL3-CK (SEQ ID NO: 3), a new Fc variant, Fv-4-F1389 comprising VH3-F1389 (SEQ ID NO: 10) and VL3-CK (SEQ ID NO: 3), were expressed and purified by the method known to those skilled in the art described in Reference Example 2 of WO2011/122011.
  • Fv-4-F1389 has a novel Fc region with improved binding affinity to human FcRn at acidic pH, but significantly reduced binding to rheumatoid factor.
  • an in vivo study of Fv-4-IgG1 and Fv-4-F1389 was performed using human FcRn transgenic mice.
  • FIG. 133 shows plasma antibody concentration time profile after injection of Fv-4-IgG1 and Fv-4-F1389.
  • Fv-4-F1389 having novel Fc variants with improved binding to FcRn at acidic pH and reduced binding to rheumatoid factor exhibited improved pharmacokinetics.
  • Novel Fc variants described in Table 28 have increased binding affinity to FcRn at pH6.0 to a same level as F1389. Therefore, these variants are also expected to exhibit improved pharmacokinetics using human FcRn transgenic mouse line 32 while having reduced binding to rheumatoid factor.
  • hIgA Human IgA
  • H (WT)-IgA1 SEQ ID NO: 12
  • L (WT) SEQ ID NO: 13
  • GA2-IgG1 (heavy chain SEQ ID NO: 14; light chain SEQ ID NO: 15) is an antibody that bind to hIgA.
  • the DNA sequences encoding heavy chain of GA2-IgG1 (SEQ ID NO: 14) and light chain of GA2-IgG1 (SEQ ID NO: 15) were inserted into animal cell expression plasmids by a method known to those skilled in the art.
  • the antibody was expressed by the method described below.
  • Cells of human fetal kidney cell-derived line FreeStyle 293-F (Invitrogen) were suspended in FreeStyle 293 Expression Medium (Invitrogen). The cell suspension was seeded into a 6-well plate (3 mL/well) at a cell density of 1.33 ⁇ 10 6 cells/ml.
  • the constructed plasmids were introduced into the cells by a lipofection method.
  • the cells were cultured for four days in a CO 2 incubator (37 degrees C., 8% CO 2 , 90 rpm).
  • the antibodies were purified from the isolated culture supernatants by a method known to those skilled in the art using rProtein A SepharoseTM Fast Flow (Amersham Biosciences).
  • the absorbance (wavelength: 280 nm) of the purified antibody solutions was measured using a spectrophotometer.
  • the antibody concentrations were determined from the measured values using the absorption coefficient calculated by the PACE method (Protein Science (1995) 4, 2411-2423).
  • the antibodies isolated as described in 8-2 were assessed for their hIgA-binding activity (dissociation constant K D (M)) using Biacore T200 (GE Healthcare).
  • Running buffers used in the measurement were 0.05% tween20/20 mmol/L ACES/150 mmol/L NaCl (pH 7.4 or 5.8) containing 3 microM or 1.2 mM CaCl 2 .
  • the antibody was allowed to bind to Sensor chip CM5 (GE Healthcare) immobilized with a suitable amount of recombinant Protein A/G (Thermo Scientific) by the amino coupling method. Then, an appropriate concentration of hIgA (described in 8-1) was injected as an analyte to allow interaction with the antibody on the sensor chip. The measurement was carried out at 37 degrees C. After the measurement, 10 mmol/L glycine-HCl (pH 1.5) was injected to regenerate the sensor chip. The dissociation constant K D (M) was calculated from the measurement result by curve fitting analysis and equilibrium parameter analysis using Biacore T200 Evaluation Software (GE Healthcare). The result and obtained sensorgrams are shown in Table 29 and FIG. 134 , respectively.
  • GA2-IgG1 bound strongly to hIgA at a Ca 2+ concentration of 1.2 mM whereas the antibody bound weakly to hIgA at a Ca 2+ concentration of 3 microM. Furthermore, at a Ca 2+ concentration of 1.2 mM, GA2-IgG1 was shown to bind to human IgA strongly at pH 7.4 but weakly at pH 5.8. More specifically, GA2-IgG1 was revealed to bind to human IgA in a pH- and calcium-dependent manner.
  • GA2-F760 (heavy chain SEQ ID NO: 16; light chain SEQ ID NO: 15) was constructed by introducing amino acid substitutions L235R and S239K into GA2-IgG1 to eliminate binding to FcgammaR.
  • GA2-F1331 (heavy chain SEQ ID NO: 17; light chain SEQ ID NO: 15) was constructed by introducing amino acid substitution G236R, M252Y, S254T, T256E, N434Y, Y436V, Q438R and S440E into GA2-F760, which binds to FcRn stronger than GA2-F760 at pH 7.4.
  • the modified antibodies were expressed by the method described above using animal expression plasmids inserted with DNA sequences encoding GA2-F1331 (heavy chain SEQ ID NO: 17; light chain SEQ ID NO: 15) and GA2-F760 (heavy chain SEQ ID NO: 16; light chain SEQ ID NO: 15) by a method known to those skilled in the art.
  • the antibody concentrations were determined after purification.
  • GA2-F760 was assessed for its binding to various mouse FcgammaR (mFcgammaRI, mFcgammaRII, mFcgammaRIII, and mFcgammaRIV). The result showed that GA2-F760 did not bind to any of the receptors.
  • hIgA and anti-hIgA antibody Pharmacokinetics of hIgA and anti-hIgA antibody was assessed after administration of hIgA (human IgA; prepared as described in Example 8) alone or in combination with an anti-hIgA antibody to human FcRn transgenic mice (B6.mFcRn ⁇ / ⁇ .hFcRn Tg line 32+/+ mouse, Jackson Laboratories; Methods Mol. Biol. (2010) 602: 93-104).
  • a mixture of hIgA and anti-hIgA antibody was administered once at a dose of 10 mL/kg via the caudal vein.
  • GA2-F760 and GA2-F1331 described above were the anti-hIgA antibodies that were used.
  • the hIgA concentration was 80 microg/mL and the anti-hIgA antibody concentration was 2.69 mg/mL. Under the conditions described above, the majority of hIgA is predicted to bind to the antibody since the anti-hIgA antibody is present in sufficient excess over hIgA.
  • Blood was collected from the mice fifteen minutes, one hour, two hours, seven hours, one day, three days, seven days and fourteen days after administration. The collected blood was immediately centrifuged for 15 minutes at 12,000 rpm and 4 degrees C. to obtain the plasma. The separated plasma was stored in a freezer at ⁇ 20 degrees C. or below until measurement.
  • Anti-hIgA antibody concentrations in mouse plasma were determined by ELISA.
  • Anti-Human IgG-immobilized plates were prepared by aliquoting Anti-Human IgG (gamma-chain specific) F(ab′)2 Fragment Antibody (SIGMA) to each well of Nunc-Immuno Plate, MaxiSorp (Nalge nunc International) and allowing the plates to stand at 4 degrees C. overnight.
  • SIGMA gamma-chain specific F(ab′)2 Fragment Antibody
  • Anti-hIgA antibody standard curve samples prepared as standard solutions at plasma concentrations of 0.5, 0.25, 0.125, 0.0625, 0.03125, 0.01563, and 0.007813 microg/mL and assay samples prepared by diluting mouse plasma samples 100-fold or more were aliquoted into the Anti-Human IgG-immobilized plates, and then the plates were incubated at 25 degrees C. for one hour.
  • Goat Anti-Human IgG (gamma-chain specific) Biotin (BIOT) Conjugate was aliquoted into each well of the plates, and then the plates were incubated at 25 degrees C. for one hour.
  • Streptavidin-PolyHRP80 (Stereospecific Detection Technologies) was added to each well of the plates, after which the plates were incubated at 25 degrees C. for one hour.
  • the chromogenic reaction using TMB One Component HRP Microwell Substrate (BioFX Laboratories) as a substrate was terminated with 1N sulfuric acid (Showa Chemical), and then the reaction mixture in each well was measured using a microplate reader to measure the absorbance at 450 nm.
  • the anti-hIgA antibody concentration in mouse plasma was calculated from the absorbance of the standard curve using analysis software SOFTmax PRO (Molecular Devices).
  • the time course of plasma antibody concentrations of GA2-F1331, and GA2-F760 in human FcRn transgenic mice, which were determined by the method described above, is shown in FIG. 135 .
  • hIgA concentrations in mouse plasma were measured by ELISA.
  • Anti-Human IgA-immobilized plates were prepared by aliquoting Goat anti-Human IgA Antibody (BETHYL) into each well of Nunc-Immuno Plate, MaxiSorp (Nalge nunc International) and allowing the plates to stand at 4 degrees C. overnight.
  • hIgA standard curve samples were prepared as standard solutions at plasma concentrations of 0.4, 0.2, 0.1, 0.05, 0.025, 0.0125, and 0.00625 microg/mL and assay samples were prepared by diluting mouse plasma samples 100-fold or more.
  • Each sample (100 microL) was mixed with 200 microL of 500 ng/mL hsIL-6R at room temperature for one hour, and then it was aliquoted at 100 microL/well into the Anti-Human IgA-immobilized plates. The resulting plates were allowed to stand at room temperature for one hour. Next, after adding Biotinylated Anti-human IL-6R Antibody (R&D) into each well of the plates, the plates were incubated at room temperature for one hour. Then, after aliquoting Streptavidin-PolyHRP80 (Stereospecific Detection Technologies) into each well of the plates, the plates were incubated at room temperature for one hour.
  • R&D Biotinylated Anti-human IL-6R Antibody
  • human IgE (heavy chain SEQ ID NO: 18; light chain SEQ ID NO: 19) (the variable region is derived from an anti-human glypican3 antibody) as an antigen was expressed using FreeStyle293 (Life Technologies). Human IgE was prepared by purifying the expressed human IgE using a conventional chromatographic method known to those skilled in the art.
  • An antibody that binds to human IgE in a pH-dependent manner was selected from a number of obtained antibodies.
  • the selected anti-human IgE antibody was expressed using human IgG1 heavy chain constant region and human light chain constant region, and then purified.
  • the produced antibody was named clone 278 (heavy chain SEQ ID NO: 20; light chain SEQ ID NO: 21).
  • Antibodies capable of dissociating from antigens within the endosome can be created not only by designing them so as to bind to antigens in a pH-dependent manner, but also by designing them so as to bind to antigens in a Ca-dependent manner.
  • clone 278 and the control Xolair (omalizumab; Novartis) whose IgE-binding activity does not depend on pH/Ca were assessed for their pH dependency and pH/Ca dependency of the human IgE (hIgE)-binding activity.
  • a chemically-synthetized peptide having a human glypican 3 protein-derived sequence (SEQ ID NO: 22) whose C-terminal Lys is biotinylated (hereinafter abbreviated as “biotinylated GPC3 peptide”) was added in an appropriate amount and immobilized onto Sensor chip SA (GE Healthcare) based on the affinity between biotin and streptavidin.
  • Human IgE was immobilized onto the chip by injecting it at an appropriate concentration so as to be trapped by the biotinylated GPC3 peptide.
  • clone 278 was injected at an appropriate concentration and allowed to interact with the human IgE on the sensor chip.
  • 278-F760 (heavy chain SEQ ID NO: 23; light chain SEQ ID NO: 21) was constructed to eliminate binding to FcgammaR. Furthermore 278-F1331 (heavy chain SEQ ID NO: 24; light chain SEQ ID NO: 21) was constructed by introducing amino acid substitution G236R, M252Y, S254T, T256E, N434Y, Y436V, Q438R and S440E into 278-F760, which binds to FcRn stronger than 278-F760 at pH 7.4.
  • the modified antibodies were expressed by the method described above using animal expression plasmids inserted with DNA sequences encoding 278-F1331 (heavy chain SEQ ID NO: 24; light chain SEQ ID NO: 21) and 278-F760 (heavy chain SEQ ID NO: 23; light chain SEQ ID NO: 21) by a method known to those skilled in the art.
  • the antibody concentrations were determined after purification.
  • hIgE(Asp6) (the variable region is derived from an anti-human glypican3 antibody), which is a human IgE for in vivo assessment consisting of a heavy chain (SEQ ID NO: 25) and a light chain (SEQ ID NO: 19), was produced by the same method as described in Example 11.
  • hIgE(Asp6) is a modified molecule resulting from asparagine-to-aspartic acid alteration at the six N-linked glycosylation sites in human IgE so that the heterogeneity in the N-linked sugar chain of human IgE is not affected by time-dependent changes in the plasma concentration of human IgE as an antigen.
  • hIgE(Asp6) and anti-human IgE antibody were assessed after administration of hIgE(Asp6) in combination with an anti-hIgE antibody (278-F760 and 278-F1331) and Sanglopor (Human normal Immunoglobulin, CSL Behring) to human FcRn transgenic mice (B6.mFcRn ⁇ / ⁇ .hFcRn Tg line 32+/+ mouse, Jackson Laboratories; Methods Mol Biol. (2010) 602: 93-104).
  • hIgE(Asp6) A mixture of hIgE(Asp6), anti-human IgE antibody and Sanglopor (the concentrations are shown in Table 31) was administered once at a dose of 10 mL/kg via the caudal vein. Under the conditions described above, hIgE(Asp6) is predicted to bind almost completely to the antibody since each antibody is present sufficiently in excess over hIgE(Asp6). Blood was collected from the mice five minutes, two hours, seven hours, one day, two days, four or five days, seven days, fourteen days, twenty-one days, and twenty-eight days after administration. The collected blood was immediately centrifuged at 15,000 rpm and 4 degrees C. for 5 minutes to obtain plasma. The separated plasma was stored in a freezer at ⁇ 20 degrees C. or below until measurement.
  • Anti-hIgE antibody concentrations in mouse plasma were determined by electrochemiluminescence (ECL) assay.
  • ECL electrochemiluminescence
  • Standard curve samples were prepared at plasma concentrations of 32, 16, 8, 4, 2, 1, 0.5, and 0.25 microgram/mL.
  • the standard curve samples and mouse plasma assay samples were aliquoted into ECL plates immobilized with hIgE(Asp6). The plates were allowed to stand at 4 degrees C. overnight. Then, Anti Rabbit Antibody (Goat), SULFO-TAG Labeled (Meso Scale Discovery) was reacted at room temperature for one hour. Immediately after Read Buffer T ( ⁇ 4) (Meso Scale Discovery) was dispensed, the measurement was performed by the Sector Imager 2400 Reader (Meso Scale Discovery). The concentration in mouse plasma was calculated from the response of the standard curve using analysis software SOFTmax PRO (Molecular Devices). A time course of the plasma antibody concentration after intravenous administration, which was determined by the method described above,
  • hIgE(Asp6) concentrations in mouse plasma were determined by ELISA. Standard curve samples were prepared at plasma concentrations of 192, 96, 48, 24, 12, 6, and 3 ng/mL. Xolair (Novartis) was added at 10 microgram/mL to the standard curve samples and mouse plasma assay samples to equalize the immune complex of hIgE(Asp6) and anti-hIgE antibody.
  • human GPC3 core protein SEQ ID NO: 26
  • anti-GPC3 antibody biotinylated with NHS-PEG4-Biotin prepared in Chugai pharmaceutical Co., Ltd.
  • Sterptavidin-PolyHRP80 Sterptavidin-PolyHRP80
  • the concentration in mouse plasma was calculated from the absorbance or luminescence intensity of the standard curve using analysis software SOFTmax PRO (Molecular Devices).
  • SOFTmax PRO Molecular Devices
  • Recombinant human IL-6 receptor as an antigen was prepared as follows.
  • a cell line constitutively expressing soluble human IL-6 receptor (hereinafter referred to as hsIL-6R) having the amino acid sequence of positions 1 to 357 from the N terminus as reported in J. Immunol. 152: 4958-4968 (1994) was established by a method known to those skilled in the art.
  • the cells were cultured to express hsIL-6R.
  • the hsIL-6R was purified from the culture supernatant by two steps: Blue Sepharose 6 FF column chromatography and gel filtration chromatography. A fraction eluted as the main peak in the final stage was used as the final purification product.
  • FcRn is a heterodimer of FcRn alpha chain and beta2-microglobulin.
  • Oligo-DNA primers were prepared based on the published human FcRn gene sequence (J Exp Med. 1994 Dec. 1; 180(6): 2377-81).
  • a DNA fragment encoding the whole gene was prepared by PCR using human cDNA (Human Placenta Marathon-Ready cDNA, Clontech) as a template and the prepared primers.
  • a DNA fragment encoding the extracellular domain containing the signal region (Met1-Leu290) was amplified by PCR, and inserted into a mammalian cell expression vector.
  • oligo-DNA primers were prepared based on the published human beta2-microglobulin gene sequence (Proc. Natl. Acad. Sci. U.S.A. 99 (26): 16899-16903 (2002)).
  • a DNA fragment encoding the whole gene was prepared by PCR using human cDNA (Human Placenta Marathon-Ready cDNA, Clontech) as a template and the prepared primers.
  • a DNA fragment encoding the whole protein containing a signal region was amplified by PCR and inserted into a mammalian cell expression vector.
  • Soluble human FcRn was expressed by the following procedure.
  • the plasmids constructed for expressing human FcRn alpha chain (SEQ ID NO: 27) and beta2-microglobulin (SEQ ID NO: 28) were introduced into cells of the human embryonic kidney cancer-derived cell line HEK293H (Invitrogen) by the lipofection method using PEI (Polyscience).
  • the resulting culture supernatant was collected, and FcRn was purified using IgG Sepharose 6 Fast Flow (Amersham Biosciences), followed by further purification using HiTrap Q HP (GE Healthcare) (J Immunol. 2002 Nov. 1; 169(9): 5171-80).
  • hIgA comprising H (WT)-IgA1 (SEQ ID NO: 12) and L (WT) (SEQ ID NO: 13) was expressed and purified by the method known to those skilled in the art using rProtein L-agarose (ACTIgen) followed by gel filtration chromatography.
  • hsPlexin A1 Recombinant soluble human plexin A1 as an antigen (hereinafter referred to as hsPlexin A1) was prepared as follows.
  • hsPlexin A1 was constructed by reference to NCBI Reference Sequence (NP_115618). Specially, hsPlexin A1 was comprised of the amino acid sequence of positions 27-1243 from the above-mentioned NCBI Reference FLAG-tag (DYKDDDDK, SEQ ID NO: 29) was connected to its C terminus.
  • hsPlexin A1 was transiently expressed using FreeStyle293 (Invitrogen) and purified from the culture supernatant by two steps: anti-FLAG column chromatography and gel filtration chromatography. A fraction eluted as the main peak in the final stage was used as the final purification product.
  • the antigen When a conventional antibody targeting soluble antigen is administered to a subject, the antigen binds to the antibody and persists stably in plasma. Since an antigen bound to an antibody has a significantly longer half-life than an antigen alone, the antigen concentration increases after the injection of a conventional antibody to approximately 10 to 1000-folds of total plasma antigen concentration from the baseline. Such an increase of the total plasma antigen concentration is not preferable for a therapeutic antibody, because the antibody concentration (dosage) has to be 10 to 1000-fold higher than necessary compared to when no substantial increase in total plasma antigen concentration occurs. Therefore, an antibody which eliminates the antigen from plasma and also reduces the total plasma antigen concentration compared to a conventional antibody is extremely valuable since the required dosage would be 10 to 1000-fold lower than that required for a conventional antibody.
  • the present inventors conducted dedicated studies on modified FcRn-binding domains which have an enhanced affinity for FcRn at neutral pH and antigen-binding molecules comprising said FcRn-binding domain which have low immunogenicity, high stability and form only a few aggregates.
  • substitutions at specific positions of the FcRn-binding domain increases the affinity for the FcRn at neutral pH without substantially increasing the immunogenicity and the ratio of high molecular weight species, and without substantially decreasing stability of antigen-binding molecules comprising the FcRn-binding domain.
  • the antigen-binding molecules comprising the FcRn-binding domain of the present invention are superior in pharmacokinetics in facilitating the reduction of the plasma antigen concentration and meet the developability criteria of low immunogenicity, high stability and very few aggregates.
  • Fc-engineering to increase the binding affinity to FcRn at neutral or acidic pH can improve the endosomal recycling efficiency and the pharmacokinetics of the antibody.
  • modifications of the amino acid sequence of an antibody e.g. amino acid substitutions and insertions
  • the present inventors conducted dedicated studies on antigen-binding molecules comprising a modified FcRn-binding domain whose binding activity for a pre-existing anti-drug antibody (ADA) was increased at neutral pH due to substitutions in the FcRn-binding domain that increased the affinity for FcRn at neutral pH or acidic pH. As a result, it was discovered that other substitutions at specific positions of the FcRn-binding domain decrease the binding activity for a pre-existing anti-drug antibody (ADA) at neutral pH while maintaining to a high extent the increased FcRn-binding activity in the respective pH ranges.
  • the antigen-binding molecules of the present invention are superior in pharmacokinetics in facilitating the reduction of the plasma antigen concentration without increasing the antibody clearance.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
US14/347,187 2011-09-30 2012-09-28 Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance Active 2035-07-17 US10253100B2 (en)

Applications Claiming Priority (16)

Application Number Priority Date Filing Date Title
JP2011218736 2011-09-30
JP2011-218736 2011-09-30
WOPCT/JP2012/058603 2012-03-30
JPPCT/JP2012/058603 2012-03-30
PCT/JP2012/058603 WO2012133782A1 (ja) 2011-03-30 2012-03-30 抗原結合分子の血漿中滞留性と免疫原性を改変する方法
JP2012-123781 2012-05-30
JP2012-123782 2012-05-30
JP2012123782 2012-05-30
JP2012-123773 2012-05-30
JP2012123781 2012-05-30
JP2012123773 2012-05-30
JP2012-139211 2012-06-20
JP2012139211 2012-06-20
JP2012-177311 2012-08-09
JP2012177311 2012-08-09
PCT/JP2012/006218 WO2013046704A2 (en) 2011-09-30 2012-09-28 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2012/006218 A-371-Of-International WO2013046704A2 (en) 2011-09-30 2012-09-28 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/264,735 Continuation US20190233525A1 (en) 2011-09-30 2019-02-01 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE

Publications (2)

Publication Number Publication Date
US20140363428A1 US20140363428A1 (en) 2014-12-11
US10253100B2 true US10253100B2 (en) 2019-04-09

Family

ID=47996560

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/347,187 Active 2035-07-17 US10253100B2 (en) 2011-09-30 2012-09-28 Therapeutic antigen-binding molecule with a FcRn-binding domain that promotes antigen clearance
US16/264,735 Abandoned US20190233525A1 (en) 2011-09-30 2019-02-01 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE
US18/533,360 Pending US20240117059A1 (en) 2011-09-30 2023-12-08 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/264,735 Abandoned US20190233525A1 (en) 2011-09-30 2019-02-01 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE
US18/533,360 Pending US20240117059A1 (en) 2011-09-30 2023-12-08 THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE

Country Status (14)

Country Link
US (3) US10253100B2 (he)
EP (3) EP4324850A2 (he)
JP (4) JP6204350B2 (he)
KR (1) KR20140069332A (he)
CN (2) CN108948197A (he)
AU (2) AU2012313670B2 (he)
BR (1) BR112014007290A2 (he)
CA (1) CA2850035A1 (he)
HK (1) HK1257946A1 (he)
MX (1) MX2014003891A (he)
RU (2) RU2014117500A (he)
SG (2) SG11201401098TA (he)
TW (3) TW201817745A (he)
WO (1) WO2013046704A2 (he)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11267877B2 (en) 2017-10-06 2022-03-08 Prothena Biosciences Limited Anti-transthyretin antibodies
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11267878B2 (en) 2015-01-28 2022-03-08 Neotope Neuroscience Limited Anti-transthyretin antibodies
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11492393B2 (en) 2016-05-02 2022-11-08 Prothena Biosciences Limited Tau immunotherapy
US11584791B2 (en) 2016-05-02 2023-02-21 Prothena Biosciences Limited Antibodies recognizing tau
US11608374B2 (en) 2017-01-30 2023-03-21 Chugai Seiyaku Kabushiki Kaisha Anti-sclerostin antibodies and methods of use
US11629185B2 (en) 2015-01-28 2023-04-18 Novo Nordisk A/S Anti-transthyretin antibodies
US11643457B2 (en) 2013-03-13 2023-05-09 Prothena Biosciences Limited Tau immunotherapy
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11858980B2 (en) 2016-08-02 2024-01-02 Visterra, Inc. Engineered polypeptides and uses thereof
US11873332B2 (en) 2017-11-29 2024-01-16 Novo Nordisk A/S Lyophilized formulation of a monoclonal antibody against transthyretin
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US11912759B2 (en) 2015-01-28 2024-02-27 Novo Nordisk A/S Anti-transthyretin antibodies
US11926659B2 (en) 2019-03-03 2024-03-12 Prothena Biosciences Limited Antibodies recognizing tau
US11958896B2 (en) 2017-05-02 2024-04-16 Prothena Biosciences Limited Antibodies recognizing tau

Families Citing this family (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102467302B1 (ko) 2007-09-26 2022-11-14 추가이 세이야쿠 가부시키가이샤 항체 정상영역 개변체
TWI440469B (zh) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
TW201817745A (zh) * 2011-09-30 2018-05-16 日商中外製藥股份有限公司 具有促進抗原清除之FcRn結合域的治療性抗原結合分子
CN110680920A (zh) 2011-09-30 2020-01-14 中外制药株式会社 诱导针对靶抗原的免疫应答的抗原结合分子
AU2013268418B2 (en) 2012-05-30 2017-12-07 Chugai Seiyaku Kabushiki Kaisha Target-tissue-specific antigen-binding molecule
EP3892638A1 (en) * 2012-05-30 2021-10-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for eliminating aggregated antigens
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
JP6668241B2 (ja) 2013-09-05 2020-03-18 アムジエン・インコーポレーテツド 予測可能で、一貫性のある、且つ再現可能な糖型特性を示すFc含有分子
RU2758952C1 (ru) 2013-09-27 2021-11-03 Чугаи Сейяку Кабусики Кайся Способ получения полипептидного гетеромультимера
WO2015083764A1 (ja) 2013-12-04 2015-06-11 中外製薬株式会社 化合物の濃度に応じて抗原結合能の変化する抗原結合分子及びそのライブラリ
NZ631007A (en) 2014-03-07 2015-10-30 Alexion Pharma Inc Anti-c5 antibodies having improved pharmacokinetics
US10780182B2 (en) 2014-04-25 2020-09-22 The Trustees Of The University Of Pennsylvania Methods and compositions for treating metastatic breast cancer and other cancers in the brain
DK3142750T3 (da) 2014-05-13 2020-09-14 Univ Pennsylvania Sammensætninger omfattende aav, som udtrykker dobbelt-antistofkonstrukter og anvendelser deraf
MA40764A (fr) 2014-09-26 2017-08-01 Chugai Pharmaceutical Co Ltd Agent thérapeutique induisant une cytotoxicité
NZ730607A (en) 2014-12-19 2022-07-01 Chugai Pharmaceutical Co Ltd Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
ES2899894T3 (es) 2014-12-19 2022-03-15 Chugai Pharmaceutical Co Ltd Anticuerpos anti-C5 y métodos de uso
MA41459A (fr) * 2015-02-03 2017-12-12 Als Therapy Development Inst Anticorps anti-cd40l et méthodes pour traiter des maladies ou des troubles liés aux cd40l
EA201791754A1 (ru) 2015-02-05 2019-01-31 Чугаи Сейяку Кабусики Кайся АНТИТЕЛА, СОДЕРЖАЩИЕ ЗАВИСЯЩИЙ ОТ КОНЦЕНТРАЦИИ ИОНОВ АНТИГЕНСВЯЗЫВАЮЩИЙ ДОМЕН, ВАРИАНТЫ Fc-ОБЛАСТИ, IL-8-СВЯЗЫВАЮЩИЕ АНТИТЕЛА И ИХ ПРИМЕНЕНИЯ
BR112017014067B1 (pt) 2015-02-27 2021-01-12 Chugai Seiyaku Kabushiki Kaisha usos de um anticorpo receptor de il-6 para no tratamento de doenças relacionadas a il-6
FR3038517B1 (fr) * 2015-07-06 2020-02-28 Laboratoire Francais Du Fractionnement Et Des Biotechnologies Utilisation de fragments fc modifies en immunotherapie
FR3039832A1 (fr) * 2015-08-04 2017-02-10 Univ Francois Rabelais De Tours Igg1 et leur utilisation therapeutique
CA2997263C (en) * 2015-09-08 2022-10-04 Theripion, Inc. Apoa-1 fusion polypeptides and related compositions and methods
EP3350202A1 (en) 2015-09-18 2018-07-25 Chugai Seiyaku Kabushiki Kaisha Il-8-binding antibodies and uses thereof
EP4316512A3 (en) 2015-10-28 2024-04-24 The Trustees of The University of Pennsylvania Intrathecal administration of adeno-associated-viral vectors for gene therapy
CN116239693A (zh) * 2016-03-14 2023-06-09 奥斯陆大学 具有改变的FcRn结合的工程化免疫球蛋白
CN109415444B (zh) 2016-07-29 2024-03-01 中外制药株式会社 显示增加的备选fviii辅因子功能活性的双特异性抗体
SG10201607778XA (en) 2016-09-16 2018-04-27 Chugai Pharmaceutical Co Ltd Anti-Dengue Virus Antibodies, Polypeptides Containing Variant Fc Regions, And Methods Of Use
CA3041279A1 (en) * 2016-11-28 2018-05-31 Chugai Seiyaku Kabushiki Kaisha Antigen-binding domain, and polypeptide including conveying section
WO2018203545A1 (ja) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Il-6及び好中球の関連する疾患の治療効果の予測及び判定方法
EP3621986A1 (en) * 2017-05-12 2020-03-18 Shire Human Genetic Therapies, Inc. Recombinant follistatin-fc fusion proteins and use in treating duchenne muscular dystrophy
RU2770209C2 (ru) * 2017-05-24 2022-04-14 ЭйЭлЭс ТЕРАПИ ДЕВЕЛОПМЕНТ ИНСТИТЬЮТ Терапевтические антитела против лиганда cd40
CN110621697B (zh) 2017-05-25 2023-06-27 百时美施贵宝公司 拮抗性cd40单克隆抗体及其用途
JP7079844B2 (ja) 2017-07-27 2022-06-02 アレクシオン ファーマシューティカルズ, インコーポレイテッド 高濃度抗c5抗体製剤
CA3072099A1 (en) * 2017-08-11 2019-02-14 Research Development Foundation Engineered antibody fc variants for enhanced serum half life
WO2019113224A1 (en) 2017-12-05 2019-06-13 The Trustees Of The University Of Pennsylvania Fusion proteins and antibodies targeting human red blood cell antigens
JP2021518343A (ja) 2018-03-15 2021-08-02 中外製薬株式会社 ジカウイルスに対して交差反応性を有する抗デングウイルス抗体および使用方法
JP7372237B2 (ja) 2018-06-04 2023-10-31 中外製薬株式会社 細胞質内での半減期が変化した抗原結合分子
AU2019318031A1 (en) 2018-08-10 2021-02-25 Chugai Seiyaku Kabushiki Kaisha Anti-CD137 antigen-binding molecule and utilization thereof
CN113194983A (zh) 2018-09-06 2021-07-30 奇达拉治疗公司 用于治疗病毒感染的组合物及方法
AU2019370339A1 (en) * 2018-11-01 2021-06-10 Shandong New Time Pharmaceutical Co., Ltd. Bispecific antibody and use thereof
AR117091A1 (es) 2018-11-19 2021-07-07 Bristol Myers Squibb Co Anticuerpos monoclonales antagonistas contra cd40 y sus usos
TW202035442A (zh) * 2018-12-20 2020-10-01 美商建南德克公司 經修飾之抗體Fc及其使用方法
EP3943108A4 (en) 2019-03-19 2023-01-04 Chugai Seiyaku Kabushiki Kaisha ANTIGEN-BINDING MOLECULE CONTAINING AN ANTIGEN-BINDING DOMAIN WHOSE ANTIGEN-BINDING ACTIVITY IS ALTERED DEPENDING ON THE MTA, AND BANK FOR OBTAINING SUCH ANTIGEN-BINDING DOMAIN
KR20210149779A (ko) 2019-04-10 2021-12-09 추가이 세이야쿠 가부시키가이샤 Fc 영역 개변 항체의 정제 방법
AU2020275348A1 (en) * 2019-05-15 2021-12-09 Chugai Seiyaku Kabushiki Kaisha An antigen-binding molecule, a pharmaceutical composition, and a method
MX2022001038A (es) * 2019-07-25 2022-04-26 Genzyme Corp Métodos para tratar trastornos mediados por anticuerpos con antagonistas de fcrn.
CA3152009A1 (en) * 2019-08-22 2021-02-25 Cidara Therapeutics, Inc. Variant fc domains and uses thereof
TW202122117A (zh) 2019-09-06 2021-06-16 美商席達拉醫療有限公司 用於治療病毒感染之組合物及方法
IL293310A (he) 2019-12-18 2022-07-01 Hoffmann La Roche נוגדני אנטי-ccl2 ביספציפיים
BR112022011723A2 (pt) * 2019-12-27 2022-09-06 Chugai Pharmaceutical Co Ltd Anticorpo anti-ctla-4 e uso do mesmo
TW202144395A (zh) 2020-02-12 2021-12-01 日商中外製藥股份有限公司 用於癌症之治療的抗cd137抗原結合分子
AR122766A1 (es) * 2020-06-29 2022-10-05 Hanall Biopharma Co Ltd Formulación para anticuerpos anti-fcrn
US20230416371A1 (en) 2020-08-28 2023-12-28 Chugai Seiyaku Kabushiki Kaisha Heterodimer fc polypeptide
CN113063949A (zh) * 2021-03-23 2021-07-02 中国医学科学院输血研究所 一种血浆中特异性IgM抗体的定量测定方法
AR125344A1 (es) 2021-04-15 2023-07-05 Chugai Pharmaceutical Co Ltd Anticuerpo anti-c1s
CN117321219A (zh) 2021-05-19 2023-12-29 中外制药株式会社 预测分子的体内药代动力学的方法
CA3221735A1 (en) 2021-06-18 2022-12-22 F. Hoffmann-La Roche Ag Bispecific anti-ccl2 antibodies
WO2022270611A1 (ja) 2021-06-25 2022-12-29 中外製薬株式会社 抗ctla-4抗体
WO2022270612A1 (ja) 2021-06-25 2022-12-29 中外製薬株式会社 抗ctla-4抗体の使用

Citations (199)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0182495A1 (en) 1984-10-15 1986-05-28 New England Medical Center Hospitals, Inc. Immunoassay means and methods useful in human native prothrombin and human abnormal prothrombin determinations
JPS6352890A (ja) 1986-08-24 1988-03-07 Chemo Sero Therapeut Res Inst 第9因子コンホメ−シヨン特異性モノクロ−ナル抗体
WO1988004692A1 (en) 1986-12-19 1988-06-30 Immunex Corporation Immunoaffinity purification system
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
WO1989001343A1 (en) 1987-08-17 1989-02-23 The Regents Of The University Of California Cationized antibodies for delivery through the blood-brain barrier
JPH01144991A (ja) 1987-12-02 1989-06-07 Kagaku Oyobi Ketsusei Riyouhou Kenkyusho 血液凝固第8因子の精製方法
JPH0228200A (ja) 1988-02-17 1990-01-30 Neorx Corp 電荷調整によるタンパク質の薬物動力学の変更
JPH02163085A (ja) 1989-11-08 1990-06-22 Teijin Ltd ヒト・プロテインcの分離方法
JPH03500664A (ja) 1987-10-27 1991-02-14 アライド‐シグナル・インコーポレーテッド ニトリルゴム及びエチレンとα‐オレフィンとのコポリマーを含有するポリアミド組成物
WO1992019759A1 (en) 1991-04-25 1992-11-12 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin 6 receptor
WO1995002187A1 (en) 1993-07-09 1995-01-19 The Institute Of Cancer Research Cell growth factor receptors
JPH0767688A (ja) 1992-09-09 1995-03-14 Immunogen Inc げっ歯類抗体の表面再処理
WO1995014710A1 (en) 1993-11-24 1995-06-01 Yeda Research And Development Co., Ltd. Chemically modified binding protein
WO1996011020A1 (fr) 1994-10-07 1996-04-18 Chugai Seiyaku Kabushiki Kaisha Medicament contre la polyarthrite rhumatoide contenant un antagoniste d'interleukine 6 comme principe actif
WO1996012503A1 (fr) 1994-10-21 1996-05-02 Chugai Seiyaku Kabushiki Kaisha Remede contre des maladies provoquees par la production d'il-6
WO1997034631A1 (en) 1996-03-18 1997-09-25 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
WO1998003546A1 (en) 1996-07-19 1998-01-29 Amgen Inc. Analogs of cationic proteins
WO1999018212A1 (fr) 1997-10-03 1999-04-15 Chugai Seiyaku Kabushiki Kaisha Anticorps humain naturel
US5935935A (en) 1993-06-10 1999-08-10 Genetic Therapy, Inc. Adenoviral vectors for treatment of hemophilia
WO1999051743A1 (fr) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Anticorps humanise contre le facteur tissulaire humain (tf) et procede de production d'anticorps humanises
WO1999058572A1 (en) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US5990286A (en) 1996-12-18 1999-11-23 Techniclone, Inc. Antibodies with reduced net positive charge
WO2000014220A1 (en) 1998-09-08 2000-03-16 Schering Aktiengesellschaft Methods and compositions for modulating the interaction between the apj receptor and the hiv virus
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
WO2001030854A2 (en) 1999-10-28 2001-05-03 Active Biotech Ab Antibody to human gastrointestinal epithelial tumor antigen related to alpha 6 beta 4 integrin
WO2001082899A2 (en) 2000-05-03 2001-11-08 Mbt Munich Biotechnology Ag Cationic diagnostic, imaging and therapeutic agents associated with activated vascular sites
US6329511B1 (en) 1998-12-01 2001-12-11 Protein Design Labs, Inc. Humanized antibodies to γ-interferon
US20020098193A1 (en) 1997-03-03 2002-07-25 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
US20020137897A1 (en) 1995-01-17 2002-09-26 Fred J. Stevens A method for altering antibody light chain interactions
US20020142374A1 (en) 1998-08-17 2002-10-03 Michael Gallo Generation of modified molecules with increased serum half-lives
US20020164339A1 (en) 1995-09-14 2002-11-07 Do Couto Fernando J.R. Recombinant peptides derived from the Mc3 anti-BA46 antibody, methods of use thereof, and methods of humanizing antibody peptides
WO2003000883A1 (en) 2001-06-22 2003-01-03 Chugai Seiyaku Kabushiki Kaisha Cell proliferation inhibitors containing anti-glypican 3 antibody
WO2003020949A2 (en) 2001-08-31 2003-03-13 The General Hospital Corporation Targeted nucleic acid constructs and uses related thereto
US20030059937A1 (en) 2000-06-16 2003-03-27 Ruben Steven M. Antibodies that immunospecifically bind BLyS
WO2003057881A1 (fr) 2001-12-28 2003-07-17 Chugai Seiyaku Kabushiki Kaisha Procede de stabilisation d'une proteine
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO2003070760A2 (en) 2002-02-20 2003-08-28 F. Hoffmann-La Roche Ag Anti-amyloid beta antibodies and their use
US20030224397A1 (en) 2002-02-11 2003-12-04 Genentech, Inc. Antibody variants with faster antigen association rates
WO2003107009A2 (en) 2002-06-12 2003-12-24 Genencor International, Inc. Methods for improving a binding characteristic of a molecule
WO2003105757A2 (en) 2002-06-12 2003-12-24 Genencor International, Inc. Methods and compositions for milieu-dependent binding of a targeted agent to a target
WO2004016740A2 (en) 2002-08-15 2004-02-26 Epitomics, Inc. Humanized rabbit antibodies
WO2004029207A2 (en) 2002-09-27 2004-04-08 Xencor Inc. Optimized fc variants and methods for their generation
WO2004035752A2 (en) 2002-10-15 2004-04-29 Protein Design Labs, Inc. ALTERATION OF FcRn BINDING AFFINITIES OR SERUM HALF-LIVES OF ANTIBODIES BY MUTAGENESIS
US20040081651A1 (en) 2001-04-13 2004-04-29 Michael Karpusas Antibodies to vla-1
WO2004039826A1 (en) 2001-11-14 2004-05-13 Centocor, Inc. Anti-il-6 antibodies, compositions, methods and uses
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040110226A1 (en) 2002-03-01 2004-06-10 Xencor Antibody optimization
US20040133357A1 (en) 2001-04-17 2004-07-08 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
WO2004068931A2 (en) 2003-02-07 2004-08-19 Protein Design Labs Inc. Amphiregulin antibodies and their use to treat cancer and psoriasis
WO2004092219A2 (en) 2003-04-10 2004-10-28 Protein Design Labs, Inc Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
WO2004096273A1 (en) 2003-04-28 2004-11-11 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
WO2004099249A2 (en) 2003-05-02 2004-11-18 Xencor, Inc. Optimized fc variants and methods for their generation
JP2005101105A (ja) 2003-09-22 2005-04-14 Canon Inc 位置決め装置、露光装置、デバイス製造方法
WO2005035756A1 (ja) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha 機能蛋白質を代替する二種特異性抗体
US6884879B1 (en) 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
WO2005037867A1 (en) 2003-10-15 2005-04-28 Pdl Biopharma, Inc. ALTERATION OF Fc-FUSION PROTEIN SERUM HALF-LIVES BY MUTAGENESIS OF POSITIONS 250, 314 AND/OR 428 OF THE HEAVY CHAIN CONSTANT REGION OF IG
US20050095243A1 (en) 2003-06-05 2005-05-05 Genentech, Inc. Combination therapy for B cell disorders
WO2005047327A2 (en) 2003-11-12 2005-05-26 Biogen Idec Ma Inc. NEONATAL Fc RECEPTOR (FcRn)-BINDING POLYPEPTIDE VARIANTS, DIMERIC Fc BINDING PROTEINS AND METHODS RELATED THERETO
WO2005059106A2 (en) 2003-12-10 2005-06-30 Medarex, Inc. Interferon alpha antibodies and their uses
US6913747B1 (en) 1999-02-12 2005-07-05 Genetics Institute, Llc Humanized immunoglobulin reactive with B7 therewith
WO2005067620A2 (en) 2004-01-09 2005-07-28 Pfizer Inc. ANTIBODIES TO MAdCAM
WO2005070963A1 (en) 2004-01-12 2005-08-04 Applied Molecular Evolution, Inc Fc region variants
WO2005077981A2 (en) 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES
US20050244403A1 (en) 2004-03-24 2005-11-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
US20050260711A1 (en) 2004-03-30 2005-11-24 Deepshikha Datta Modulating pH-sensitive binding using non-natural amino acids
US20050260213A1 (en) 2004-04-16 2005-11-24 Scott Koenig Fcgamma-RIIB-specific antibodies and methods of use thereof
US20050261229A1 (en) 1999-07-21 2005-11-24 Emd Lexigen Research Center Corp. Fc fusion proteins for enhancing the immunogenicity of protein and peptide antigens
WO2005112564A2 (en) 2004-04-15 2005-12-01 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Germline and sequence variants of humanized antibodies and methods of making and using them
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
WO2005123126A2 (en) 2004-06-09 2005-12-29 Wyeth Antibodies against human interleukin-13 and uses therefor
WO2006004663A2 (en) 2004-06-25 2006-01-12 Medimmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
US20060014156A1 (en) 2001-06-30 2006-01-19 Enzo Life Sciences, Inc. Nucleic acid detecting or quantifying processes
RU2005112742A (ru) 2002-09-27 2006-01-20 Ксенкор, Инк. (Us) Оптимизированные варианты fc и способы их создания
WO2006016644A1 (ja) 2004-08-11 2006-02-16 Mitsubishi Chemical Corporation 抗体及びその利用
WO2006019447A1 (en) 2004-07-15 2006-02-23 Xencor, Inc. Optimized fc variants
WO2006020114A2 (en) 2004-08-04 2006-02-23 Applied Molecular Evolution, Inc. Variant fc regions
WO2006023403A2 (en) 2004-08-16 2006-03-02 Medimmune, Inc. Eph receptor fc variants with enhanced antibody dependent cell-mediated cytotoxicity activity
WO2006030220A1 (en) 2004-09-17 2006-03-23 Domantis Limited Compositions monovalent for cd40l binding and methods of use
WO2006031370A2 (en) 2004-08-19 2006-03-23 Genentech, Inc. Polypeptide variants with altered effector function
WO2006030200A1 (en) 2004-09-14 2006-03-23 National Institute For Biological Standards And Control Vaccine
WO2006050166A2 (en) 2004-10-29 2006-05-11 Medimmune, Inc. Methods of preventing and treating rsv infections and related conditions
WO2006050491A2 (en) 2004-11-08 2006-05-11 Epitomics, Inc. Methods for antibody engineering
WO2006053301A2 (en) 2004-11-12 2006-05-18 Xencor, Inc. Fc variants with altered binding to fcrn
US20060134709A1 (en) 2004-11-10 2006-06-22 Jeffery Stavenhagen Engineering Fc antibody regions to confer effector function
WO2006067913A1 (ja) 2004-12-22 2006-06-29 Chugai Seiyaku Kabushiki Kaisha フコーストランスポーターの機能が阻害された細胞を用いた抗体の作製方法
WO2006066598A2 (en) 2004-12-23 2006-06-29 Novo Nordisk A/S Antibody binding affinity ligands
US20060153860A1 (en) 2004-06-18 2006-07-13 Ambrx, Inc. Novel antigen-binding polypeptides and their uses
WO2006105338A2 (en) 2005-03-31 2006-10-05 Xencor, Inc. Fc VARIANTS WITH OPTIMIZED PROPERTIES
WO2006106905A1 (ja) 2005-03-31 2006-10-12 Chugai Seiyaku Kabushiki Kaisha 会合制御によるポリペプチド製造方法
WO2006109592A1 (ja) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha 血液凝固第viii因子の機能代替抗体
WO2006116260A2 (en) 2005-04-26 2006-11-02 Medimmune, Inc. Modulation of antibody effector function by hinge domain engineering
WO2006121852A2 (en) 2005-05-05 2006-11-16 Duke University Anti-cd19 antibody therapy for autoimmune disease
WO2006130834A2 (en) 2005-05-31 2006-12-07 Board Of Regents, The University Of Texas System IGGl ANTIBODIES WITH MUTATED FC PORTION FOR INCREASED BINDING TO FCRN RECEPTOR AND USES THEREOF
US20070009523A1 (en) * 1999-01-15 2007-01-11 Genentech, Inc. Polypeptide variants with altered effector function
US20070037734A1 (en) 2003-11-05 2007-02-15 Ares Trading S.A. Process for the purification of il-18 binding protein
WO2007021841A2 (en) 2005-08-10 2007-02-22 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
WO2007024535A2 (en) 2005-08-19 2007-03-01 Wyeth Antagonist antibodies against gdf-8 and uses in treatment of als and other gdf-8-associated disorders
US20070059312A1 (en) 1997-04-07 2007-03-15 Genentech, Inc. Anti-vegf antibodies
WO2007041635A2 (en) 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
EP1601697B1 (en) 2003-02-28 2007-05-30 Lonza Biologics plc Antibody purification by Protein A and ion exchange chromatography
WO2007060411A1 (en) 2005-11-24 2007-05-31 Ucb Pharma S.A. Anti-tnf alpha antibodies which selectively inhibit tnf alpha signalling through the p55r
WO2007076524A2 (en) 2005-12-29 2007-07-05 Centocor, Inc. Human anti-il-23 antibodies, compositions, methods and uses
US20070160598A1 (en) 2005-11-07 2007-07-12 Dennis Mark S Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
US20070231329A1 (en) 2003-03-03 2007-10-04 Xencor, Inc. Fc Variants Having Increased Affinity for FcyRIIb
WO2007114319A1 (ja) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha 抗体の血中動態を制御する方法
WO2007114325A1 (ja) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha 二重特異性抗体を精製するための抗体改変方法
US20070253951A1 (en) 2006-04-24 2007-11-01 Gordon Ng Humanized c-Kit antibody
US20070269371A1 (en) 2003-09-05 2007-11-22 Genentech, Inc. Antibodies with altered effector functions
WO2007142325A1 (ja) 2006-06-08 2007-12-13 Chugai Seiyaku Kabushiki Kaisha 炎症性疾患の予防または治療剤
CN101098890A (zh) 2004-11-12 2008-01-02 赞科股份有限公司 对FcRn的结合被改变的Fc变体
WO2008002933A2 (en) 2006-06-26 2008-01-03 Macrogenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
US20080044417A1 (en) 2006-05-26 2008-02-21 Macrogenics, Inc. Humanized Fc.gamma.RIIB-Specific Antibodies and Methods of Use Thereof
WO2008022152A2 (en) 2006-08-14 2008-02-21 Xencor, Inc. Optimized antibodies that target cd19
WO2008043822A2 (en) 2006-10-11 2008-04-17 Ablynx N. V. Amino acid sequences that bind to a desired molecule in a conditional manner
WO2008060785A2 (en) 2006-10-06 2008-05-22 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Prevention of tissue ischemia, related methods and compositions
US20080138349A1 (en) 2006-12-08 2008-06-12 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2008091954A2 (en) 2007-01-23 2008-07-31 Xencor, Inc. Optimized cd40 antibodies and methods of using the same
WO2008092117A2 (en) 2007-01-25 2008-07-31 Xencor, Inc. Immunoglobulins with modifications in the fcr binding region
US20080199471A1 (en) 2002-03-01 2008-08-21 Bernett Matthew J Optimized cd40 antibodies and methods of using the same
RU2337107C2 (ru) 2003-05-02 2008-10-27 Ксенкор, Инк. ОПТИМИЗИРОВАННЫЕ Fc-ВАРИАНТЫ, ИМЕЮЩИЕ ИЗМЕНЕННОЕ СВЯЗЫВАНИЕ С FcγR, И СПОСОБЫ ИХ ПОЛУЧЕНИЯ
WO2008143954A2 (en) 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
US20080292637A1 (en) 2005-11-30 2008-11-27 Pnina Fishman Therapeutic Uses of A3 Adenosine Receptor Antibodies
WO2009006338A1 (en) 2007-06-29 2009-01-08 Quest Diagnostics Investments Incorporated Analysis of amino acids in body fluid by liquid chromatography-mass spectrometry
WO2009008529A1 (ja) 2007-07-06 2009-01-15 Tokyo Metropolitan Organization For Medical Research Tdp-43凝集物に特異的に結合する抗体
US20090041770A1 (en) 2004-11-12 2009-02-12 Chamberlain Aaron Keith Fc VARIANTS WITH ALTERED BINDING TO FcRn
US20090053240A1 (en) 2004-10-21 2009-02-26 Xencor, Inc. Novel Immunoglobulin Insertions, Deletions and Substitutions
US20090076251A1 (en) 2002-08-14 2009-03-19 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
WO2009041643A1 (ja) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Cdrのアミノ酸置換により抗体の等電点を改変する方法
CA2700986A1 (en) 2007-09-28 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
WO2009053358A1 (en) 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying fc-fusion proteins
WO2009062083A2 (en) 2007-11-08 2009-05-14 Pikamab, Inc. Methods and compositions for antibody therapy
US20090136485A1 (en) 2007-05-30 2009-05-28 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
US20090142340A1 (en) 2002-03-01 2009-06-04 Xencor, Inc. Optimized Fc Variants and Methods for Their Generation
WO2009086320A1 (en) 2007-12-26 2009-07-09 Xencor, Inc Fc variants with altered binding to fcrn
WO2009095235A1 (en) 2008-01-29 2009-08-06 Ablynx N.V. Methods to stabilize proteins and polypeptides
US20090215991A1 (en) 2003-03-03 2009-08-27 Xencor, Inc. Optimized Fc Variants and methods for their generation
WO2009125825A1 (ja) 2008-04-11 2009-10-15 中外製薬株式会社 複数分子の抗原に繰り返し結合する抗原結合分子
WO2009139822A1 (en) 2008-05-01 2009-11-19 Amgen Inc. Anti-hepcidin antibodies and methods of use
WO2009155513A2 (en) 2008-06-20 2009-12-23 Novartis Ag Immunoglobulins with reduced aggregation
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
JP2010079667A (ja) 2008-09-26 2010-04-08 Kyocera Mita Corp 画像処理装置、画像処理方法及び画像処理プログラム
US20100098730A1 (en) 2008-10-14 2010-04-22 Lowman Henry B Immunoglobulin variants and uses thereof
WO2010058860A1 (ja) 2008-11-18 2010-05-27 株式会社シノテスト 試料中のc反応性蛋白質の測定方法及び測定試薬
WO2010081173A2 (en) 2009-01-12 2010-07-15 Cytomx Therapeutics, Llc Modified antibody compositions, methods of making and using thereof
US20100184959A1 (en) 2007-03-19 2010-07-22 Medimmune Limited Polypeptide Variants
WO2010085682A2 (en) 2009-01-23 2010-07-29 Biogen Idec Ma Inc. Stabilized fc polypeptides with reduced effector function and methods of use
WO2010107109A1 (ja) 2009-03-19 2010-09-23 中外製薬株式会社 抗体定常領域改変体
WO2010106180A2 (en) 2009-03-20 2010-09-23 Lfb Biotechnologies Optimized fc variants
JP2010250830A (ja) 2009-04-16 2010-11-04 Accenture Global Services Gmbh ウェブ・サイト・アクセラレータ
US20100292443A1 (en) 2009-02-26 2010-11-18 Sabbadini Roger A Humanized platelet activating factor antibody design using anti-lipid antibody templates
US20100298542A1 (en) 2007-09-26 2010-11-25 Chugai Seiyaku Kabushiki Kaisha Modified Antibody Constant Region
US20110044986A1 (en) 2007-12-21 2011-02-24 Amgen Inc. Anti-amyloid antibodies and uses thereof
WO2011044368A1 (en) 2009-10-07 2011-04-14 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2011043643A1 (en) 2009-10-06 2011-04-14 Medimmune Ltd Rsv-specific binding molecule
US20110098450A1 (en) 2008-09-26 2011-04-28 Chugai Seiyaku Kabushiki Kaisha Antibody Molecules
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2011094593A2 (en) 2010-01-28 2011-08-04 Ab Biosciences, Inc. Novel lowered affinity antibodies and methods of marking the same
US20110223658A1 (en) 2000-04-12 2011-09-15 Lfb Biotechnologies Monoclonal antibodies with enhanced adcc function
WO2011111007A2 (en) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation ANTIBODIES WITH pH DEPENDENT ANTIGEN BINDING
JP2011184418A (ja) 2010-03-11 2011-09-22 Tokyo Institute Of Technology 親和性可変抗体
CA2794860A1 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
WO2011122011A2 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
US20110245473A1 (en) 2007-09-26 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Anti-IL-6 Receptor Antibody
WO2012033953A1 (en) 2010-09-08 2012-03-15 Halozyme, Inc. Methods for assessing and identifying or evolving conditionally active therapeutic proteins
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
WO2012073992A1 (ja) 2010-11-30 2012-06-07 中外製薬株式会社 複数分子の抗原に繰り返し結合する抗原結合分子
CA2827923A1 (en) 2011-02-25 2012-08-30 Chugai Seiyaku Kabushiki Kaisha Fc.gamma.riib-specific fc antibody
WO2012133782A1 (ja) 2011-03-30 2012-10-04 中外製薬株式会社 抗原結合分子の血漿中滞留性と免疫原性を改変する方法
CA2831770A1 (en) 2011-03-30 2012-10-04 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US8329867B2 (en) 2010-02-19 2012-12-11 Xencor, Inc. CTLA4-Ig immunoadhesins
WO2013004842A2 (en) 2011-07-06 2013-01-10 Genmab A/S Antibody variants and uses thereof
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
WO2013047748A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 複数の生理活性を有する抗原の消失を促進する抗原結合分子
WO2013047752A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 抗原の消失を促進する抗原結合分子
WO2013047729A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 標的抗原に対する免疫応答を誘導する抗原結合分子
WO2013081143A1 (ja) 2011-11-30 2013-06-06 中外製薬株式会社 免疫複合体を形成する細胞内への運搬体(キャリア)を含む医薬
WO2013125667A1 (ja) 2012-02-24 2013-08-29 中外製薬株式会社 FcγRIIBを介して抗原の消失を促進する抗原結合分子
US8568726B2 (en) * 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
WO2013180201A1 (ja) 2012-05-30 2013-12-05 中外製薬株式会社 会合化した抗原を消失させる抗原結合分子
WO2013180200A1 (ja) 2012-05-30 2013-12-05 中外製薬株式会社 標的組織特異的抗原結合分子
WO2014028354A1 (en) 2012-08-13 2014-02-20 Regeneron Pharmaceuticals, Inc. Anti-pcsk9 antibodies with ph-dependent binding characteristics
WO2014030728A1 (ja) 2012-08-24 2014-02-27 中外製薬株式会社 FcγRIIb特異的Fc領域改変体
US20140086916A1 (en) 2011-05-25 2014-03-27 Dongxing Zha METHOD FOR PREPARING Fc CONTAINING POLYPEPTIDES HAVING IMPROVED PROPERTIES
US20140112926A1 (en) 2011-03-16 2014-04-24 Amgen Inc. Fc VARIANTS
WO2014144575A1 (en) 2013-03-15 2014-09-18 Forta Corporation Reinforcement fiber coating compositions, methods of making and treating, and uses for improved adhesion to asphalt and portland cement concrete
WO2014144080A2 (en) 2013-03-15 2014-09-18 Amgen Inc. Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9
WO2014163101A1 (ja) 2013-04-02 2014-10-09 中外製薬株式会社 Fc領域改変体
US9079949B1 (en) 2014-03-07 2015-07-14 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
US20150203577A1 (en) 2012-08-24 2015-07-23 Chugai Seiyaku Kabushiki Kaisha MOUSE FcgammaRII-SPECIFIC Fc ANTIBODY
US20150210763A1 (en) 2012-02-09 2015-07-30 Chugai Seiyaku Kabushiki Kaisha MODIFIED Fc REGION OF ANTIBODY
US20150247849A1 (en) 2012-09-21 2015-09-03 Paul P. Tamburini Screening assays for complement component c5 antagonists
US20150299313A1 (en) 2011-10-05 2015-10-22 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting clearance from plasma of antigen comprising suger chain receptor-binding domain
WO2016000813A1 (en) 2014-06-30 2016-01-07 Merck Patent Gmbh Anti-tnfa antibodies with ph-dependent antigen binding
US20160068592A1 (en) 2013-01-31 2016-03-10 Seoul National University R&Db Foundation C5 antibody and method for preventing and treating complement-related diseases
US9765135B2 (en) * 2014-12-19 2017-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-C5 antibodies
US9969800B2 (en) * 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10024867B2 (en) * 2011-09-30 2018-07-17 Chugai Seiyaku Kabushiki Kaisha Ion concentration-dependent binding molecule library

Family Cites Families (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS58201994A (ja) 1982-05-21 1983-11-25 Hideaki Hagiwara 抗原特異的ヒト免疫グロブリンの生産方法
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
DK0585287T3 (da) 1990-07-10 2000-04-17 Cambridge Antibody Tech Fremgangsmåde til fremstilling af specifikke bindingsparelementer
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
WO1993012227A1 (en) 1991-12-17 1993-06-24 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
DK0546073T3 (da) 1990-08-29 1998-02-02 Genpharm Int Frembringelse og anvendelse af transgene, ikke-humane dyr, der er i stand til at danne heterologe antistoffer
EP0605522B1 (en) 1991-09-23 1999-06-23 Medical Research Council Methods for the production of humanized antibodies
WO1993011236A1 (en) 1991-12-02 1993-06-10 Medical Research Council Production of anti-self antibodies from antibody segment repertoires and displayed on phage
EP0656941B1 (en) 1992-03-24 2005-06-01 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1994002602A1 (en) 1992-07-24 1994-02-03 Cell Genesys, Inc. Generation of xenogeneic antibodies
GB9313509D0 (en) 1993-06-30 1993-08-11 Medical Res Council Chemisynthetic libraries
FR2707189B1 (fr) 1993-07-09 1995-10-13 Gradient Ass Procédé de traitement de résidus de combustion et installation de mise en Óoeuvre dudit procédé.
US6214613B1 (en) 1993-12-03 2001-04-10 Ashai Kasei Kogyo Kabushiki Kaisha Expression screening vector
EP0731842A1 (en) 1993-12-03 1996-09-18 Medical Research Council Recombinant binding proteins and peptides
WO1996002576A1 (fr) 1994-07-13 1996-02-01 Chugai Seiyaku Kabushiki Kaisha Anticorps humain reconstitue contre l'interleukine-8 humaine
DE69637481T2 (de) 1995-04-27 2009-04-09 Amgen Fremont Inc. Aus immunisierten Xenomäusen stammende menschliche Antikörper gegen IL-8
AU2466895A (en) 1995-04-28 1996-11-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
CZ106299A3 (cs) 1996-09-26 1999-08-11 Chugai Seiyaku Kabushiki Kaisha Protilátky proti proteinu příbuznému s lidským parathyroidálním hormonem
FR2761994B1 (fr) 1997-04-11 1999-06-18 Centre Nat Rech Scient Preparation de recepteurs membranaires a partir de baculovirus extracellulaires
US5897781A (en) 1997-06-06 1999-04-27 Waters Investments Limited Active pump phasing to enhance chromatographic reproducibility
ES2420835T3 (es) 1999-04-09 2013-08-27 Kyowa Hakko Kirin Co., Ltd. Procedimiento para controlar la actividad de las moléculas inmunofuncionales
AU2002218166A1 (en) 2000-09-08 2002-03-22 Universitat Zurich Collections of repeat proteins comprising repeat modules
CN103333860B (zh) 2000-10-06 2015-07-08 协和发酵麒麟株式会社 产生抗体组合物的细胞
AU1325102A (en) 2000-10-16 2002-04-29 Phylos Inc Protein scaffolds for antibody mimics and other binding proteins
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
JP2005500018A (ja) 2001-04-02 2005-01-06 アイデック ファーマスーティカルズ コーポレイション GnTIIIと同時発現する組換え抗体
US20030157561A1 (en) 2001-11-19 2003-08-21 Kolkman Joost A. Combinatorial libraries of monomer domains
DE60237282D1 (de) 2001-06-28 2010-09-23 Domantis Ltd Doppelspezifischer ligand und dessen verwendung
AU2003290330A1 (en) 2002-12-27 2004-07-22 Domantis Limited Dual specific single domain antibodies specific for a ligand and for the receptor of the ligand
NZ544486A (en) 2003-06-13 2009-04-30 Biogen Idec Inc Aglycosyl anti-cd154 (cd40 ligand) antibodies and uses thereof
US20050164301A1 (en) 2003-10-24 2005-07-28 Avidia Research Institute LDL receptor class A and EGF domain monomers and multimers
DE102004032634A1 (de) 2004-07-06 2006-02-16 Sms Demag Ag Verfahren und Einrichtung zum Messen und Regeln der Planheit und/oder der Bandspannungen eines Edelstahlbandes oder einer Edelstahlfolie beim Kaltwalzen in einem Vielwalzengerüst, insbesondere in einem 20-Walzen-Sendizimir-Walzwerk
EP1988922A4 (en) 2006-02-03 2010-06-02 Medimmune Llc PROTEIN FORMULATIONS
CA2764852C (en) 2007-01-05 2018-09-18 University Of Zurich Method of providing disease-specific binding molecules and targets
EP2144931A2 (en) 2007-04-04 2010-01-20 The Government Of The U.S.A, As Represented By The Secretary, Dept. Of Health And Human Services Monoclonal antibodies against dengue and other viruses with deletion in fc region
TW201817745A (zh) * 2011-09-30 2018-05-16 日商中外製藥股份有限公司 具有促進抗原清除之FcRn結合域的治療性抗原結合分子

Patent Citations (327)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS61117457A (ja) 1984-10-15 1986-06-04 ニユ−・イングランド・メデイカル・センタ−・ホスピタルズ・インコ−ポレ−テツド モノクロ−ナル抗体
EP0182495A1 (en) 1984-10-15 1986-05-28 New England Medical Center Hospitals, Inc. Immunoassay means and methods useful in human native prothrombin and human abnormal prothrombin determinations
JPS6352890A (ja) 1986-08-24 1988-03-07 Chemo Sero Therapeut Res Inst 第9因子コンホメ−シヨン特異性モノクロ−ナル抗体
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
JPH02501112A (ja) 1986-12-19 1990-04-19 イーストマン・コダック・カンパニー 免疫親和性による精製方法
WO1988004692A1 (en) 1986-12-19 1988-06-30 Immunex Corporation Immunoaffinity purification system
WO1989001343A1 (en) 1987-08-17 1989-02-23 The Regents Of The University Of California Cationized antibodies for delivery through the blood-brain barrier
JPH03500664A (ja) 1987-10-27 1991-02-14 アライド‐シグナル・インコーポレーテッド ニトリルゴム及びエチレンとα‐オレフィンとのコポリマーを含有するポリアミド組成物
JPH01144991A (ja) 1987-12-02 1989-06-07 Kagaku Oyobi Ketsusei Riyouhou Kenkyusho 血液凝固第8因子の精製方法
JPH0228200A (ja) 1988-02-17 1990-01-30 Neorx Corp 電荷調整によるタンパク質の薬物動力学の変更
EP0329185B1 (en) 1988-02-17 1994-04-20 Neorx Corporation Alteration of pharmacokinetics of proteins by charge modification
US5322678A (en) 1988-02-17 1994-06-21 Neorx Corporation Alteration of pharmacokinetics of proteins by charge modification
JPH02163085A (ja) 1989-11-08 1990-06-22 Teijin Ltd ヒト・プロテインcの分離方法
US5795965A (en) 1991-04-25 1998-08-18 Chugai Seiyaku Kabushiki Kaisha Reshaped human to human interleukin-6 receptor
WO1992019759A1 (en) 1991-04-25 1992-11-12 Chugai Seiyaku Kabushiki Kaisha Reconstituted human antibody against human interleukin 6 receptor
JPH0767688A (ja) 1992-09-09 1995-03-14 Immunogen Inc げっ歯類抗体の表面再処理
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5935935A (en) 1993-06-10 1999-08-10 Genetic Therapy, Inc. Adenoviral vectors for treatment of hemophilia
WO1995002187A1 (en) 1993-07-09 1995-01-19 The Institute Of Cancer Research Cell growth factor receptors
WO1995014710A1 (en) 1993-11-24 1995-06-01 Yeda Research And Development Co., Ltd. Chemically modified binding protein
US6074642A (en) 1994-05-02 2000-06-13 Alexion Pharmaceuticals, Inc. Use of antibodies specific to human complement component C5 for the treatment of glomerulonephritis
EP0783893A1 (en) 1994-10-07 1997-07-16 Chugai Seiyaku Kabushiki Kaisha Rheumatoid arthritis remedy containing il-6 antagonist as active ingredient
WO1996011020A1 (fr) 1994-10-07 1996-04-18 Chugai Seiyaku Kabushiki Kaisha Medicament contre la polyarthrite rhumatoide contenant un antagoniste d'interleukine 6 comme principe actif
WO1996012503A1 (fr) 1994-10-21 1996-05-02 Chugai Seiyaku Kabushiki Kaisha Remede contre des maladies provoquees par la production d'il-6
US20070036785A1 (en) 1994-10-21 2007-02-15 Tadamitsu Kishimoto Pharmaceutical composition for treatment of diseases caused by IL-6 production
US6485943B2 (en) 1995-01-17 2002-11-26 The University Of Chicago Method for altering antibody light chain interactions
US20020137897A1 (en) 1995-01-17 2002-09-26 Fred J. Stevens A method for altering antibody light chain interactions
US20020164339A1 (en) 1995-09-14 2002-11-07 Do Couto Fernando J.R. Recombinant peptides derived from the Mc3 anti-BA46 antibody, methods of use thereof, and methods of humanizing antibody peptides
WO1997034631A1 (en) 1996-03-18 1997-09-25 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
WO1998003546A1 (en) 1996-07-19 1998-01-29 Amgen Inc. Analogs of cationic proteins
US5990286A (en) 1996-12-18 1999-11-23 Techniclone, Inc. Antibodies with reduced net positive charge
US20020098193A1 (en) 1997-03-03 2002-07-25 Board Of Regents, The University Of Texas System Immunoglobin-like domains with increased half lives
US20070059312A1 (en) 1997-04-07 2007-03-15 Genentech, Inc. Anti-vegf antibodies
US6884879B1 (en) 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
WO1999018212A1 (fr) 1997-10-03 1999-04-15 Chugai Seiyaku Kabushiki Kaisha Anticorps humain naturel
US20030103970A1 (en) 1997-10-03 2003-06-05 Masayuki Tsuchiya Natural human antibody
US7052873B2 (en) 1997-10-03 2006-05-30 Chugai Seiyaku Kabushiki Kaisha Natural human antibody
WO1999051743A1 (fr) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Anticorps humanise contre le facteur tissulaire humain (tf) et procede de production d'anticorps humanises
EP1069185A1 (en) 1998-04-03 2001-01-17 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
US6677436B1 (en) 1998-04-03 2004-01-13 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (TF) and process of production of the humanized antibody
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO1999058572A1 (en) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US20020142374A1 (en) 1998-08-17 2002-10-03 Michael Gallo Generation of modified molecules with increased serum half-lives
WO2000014220A1 (en) 1998-09-08 2000-03-16 Schering Aktiengesellschaft Methods and compositions for modulating the interaction between the apj receptor and the hiv virus
RU2225721C2 (ru) 1998-09-08 2004-03-20 Шеринг Акциенгезельшафт Способы и композиции для модуляции взаимодействия между apj-рецептором и вирусом иммунодефицита человека (вич)
US6329511B1 (en) 1998-12-01 2001-12-11 Protein Design Labs, Inc. Humanized antibodies to γ-interferon
JP2003512019A (ja) 1999-01-15 2003-04-02 ジェネンテック・インコーポレーテッド 変化したエフェクター機能を有するポリペプチド変異体
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20070009523A1 (en) * 1999-01-15 2007-01-11 Genentech, Inc. Polypeptide variants with altered effector function
CN1763097A (zh) 1999-01-15 2006-04-26 杰南技术公司 具有改变的效应功能的多肽变体
EP2366713A2 (en) 1999-01-15 2011-09-21 Genentech, Inc. Polypeptide variants with altered effector function
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US6913747B1 (en) 1999-02-12 2005-07-05 Genetics Institute, Llc Humanized immunoglobulin reactive with B7 therewith
US20050261229A1 (en) 1999-07-21 2005-11-24 Emd Lexigen Research Center Corp. Fc fusion proteins for enhancing the immunogenicity of protein and peptide antigens
WO2001030854A2 (en) 1999-10-28 2001-05-03 Active Biotech Ab Antibody to human gastrointestinal epithelial tumor antigen related to alpha 6 beta 4 integrin
RU2266298C2 (ru) 1999-10-28 2005-12-20 Эктив Байотек Аб Антитела к антигену эпителиальных опухолей желудочно-кишечного тракта человека, родственному альфа 6 бета 4 интегрину
US20110223658A1 (en) 2000-04-12 2011-09-15 Lfb Biotechnologies Monoclonal antibodies with enhanced adcc function
JP2004511426A (ja) 2000-05-03 2004-04-15 ミュンヘン バイオテク アーゲー 活性化血管部位に関連する陽イオン性の診断薬、画像化剤、および治療薬
WO2001082899A2 (en) 2000-05-03 2001-11-08 Mbt Munich Biotechnology Ag Cationic diagnostic, imaging and therapeutic agents associated with activated vascular sites
US20030059937A1 (en) 2000-06-16 2003-03-27 Ruben Steven M. Antibodies that immunospecifically bind BLyS
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
US20040081651A1 (en) 2001-04-13 2004-04-29 Michael Karpusas Antibodies to vla-1
US20040133357A1 (en) 2001-04-17 2004-07-08 Abmaxis, Inc. Humanized antibodies against vascular endothelial growth factor
US20040236080A1 (en) 2001-06-22 2004-11-25 Hiroyuki Aburatani Cell proliferation inhibitors containing anti-glypican 3 antibody
WO2003000883A1 (en) 2001-06-22 2003-01-03 Chugai Seiyaku Kabushiki Kaisha Cell proliferation inhibitors containing anti-glypican 3 antibody
US20060014156A1 (en) 2001-06-30 2006-01-19 Enzo Life Sciences, Inc. Nucleic acid detecting or quantifying processes
WO2003020949A2 (en) 2001-08-31 2003-03-13 The General Hospital Corporation Targeted nucleic acid constructs and uses related thereto
WO2004039826A1 (en) 2001-11-14 2004-05-13 Centocor, Inc. Anti-il-6 antibodies, compositions, methods and uses
WO2003057881A1 (fr) 2001-12-28 2003-07-17 Chugai Seiyaku Kabushiki Kaisha Procede de stabilisation d'une proteine
US20030224397A1 (en) 2002-02-11 2003-12-04 Genentech, Inc. Antibody variants with faster antigen association rates
WO2003070760A2 (en) 2002-02-20 2003-08-28 F. Hoffmann-La Roche Ag Anti-amyloid beta antibodies and their use
US20040110226A1 (en) 2002-03-01 2004-06-10 Xencor Antibody optimization
US20090142340A1 (en) 2002-03-01 2009-06-04 Xencor, Inc. Optimized Fc Variants and Methods for Their Generation
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US20080199471A1 (en) 2002-03-01 2008-08-21 Bernett Matthew J Optimized cd40 antibodies and methods of using the same
WO2003105757A2 (en) 2002-06-12 2003-12-24 Genencor International, Inc. Methods and compositions for milieu-dependent binding of a targeted agent to a target
WO2003107009A2 (en) 2002-06-12 2003-12-24 Genencor International, Inc. Methods for improving a binding characteristic of a molecule
US20060141456A1 (en) 2002-06-12 2006-06-29 Cynthia Edwards Methods and compositions for milieu-dependent binding of a targeted agent to a target
US20090076251A1 (en) 2002-08-14 2009-03-19 Macrogenics, Inc. FcGammaRIIB Specific Antibodies and Methods of Use Thereof
JP2005535341A (ja) 2002-08-15 2005-11-24 エピトミスク インコーポレーティッド ヒト化ウサギ抗体
WO2004016740A2 (en) 2002-08-15 2004-02-26 Epitomics, Inc. Humanized rabbit antibodies
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
RU2005112742A (ru) 2002-09-27 2006-01-20 Ксенкор, Инк. (Us) Оптимизированные варианты fc и способы их создания
RU2390527C2 (ru) 2002-09-27 2010-05-27 Ксенкор, Инк. АНТИТЕЛО, СОДЕРЖАЩЕЕ Fc-ВАРИАНТНУЮ ЧАСТЬ (ВАРИАНТЫ), ФАРМАЦЕВТИЧЕСКАЯ КОМПОЗИЦИЯ, СОДЕРЖАЩАЯ АНТИТЕЛО, И СПОСОБ ЛЕЧЕНИЯ МЛЕКОПИТАЮЩЕГО
WO2004029207A2 (en) 2002-09-27 2004-04-08 Xencor Inc. Optimized fc variants and methods for their generation
WO2004035752A2 (en) 2002-10-15 2004-04-29 Protein Design Labs, Inc. ALTERATION OF FcRn BINDING AFFINITIES OR SERUM HALF-LIVES OF ANTIBODIES BY MUTAGENESIS
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2004068931A2 (en) 2003-02-07 2004-08-19 Protein Design Labs Inc. Amphiregulin antibodies and their use to treat cancer and psoriasis
EP1601697B1 (en) 2003-02-28 2007-05-30 Lonza Biologics plc Antibody purification by Protein A and ion exchange chromatography
US20070237767A1 (en) 2003-03-03 2007-10-11 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRllla
US8735545B2 (en) 2003-03-03 2014-05-27 Xencor, Inc. Fc variants having increased affinity for fcyrllc
US20090215991A1 (en) 2003-03-03 2009-08-27 Xencor, Inc. Optimized Fc Variants and methods for their generation
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US20070231329A1 (en) 2003-03-03 2007-10-04 Xencor, Inc. Fc Variants Having Increased Affinity for FcyRIIb
US20070248602A1 (en) 2003-03-03 2007-10-25 Xencor, Inc. Fc Variants Having Increased Affinity for FcyRllc
WO2004092219A2 (en) 2003-04-10 2004-10-28 Protein Design Labs, Inc Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
WO2004096273A1 (en) 2003-04-28 2004-11-11 Chugai Seiyaku Kabushiki Kaisha Methods for treating interleukin-6 related diseases
EP2368911A1 (en) 2003-05-02 2011-09-28 Xencor Inc. Optimized Fc variants and methods for their generation
WO2004099249A2 (en) 2003-05-02 2004-11-18 Xencor, Inc. Optimized fc variants and methods for their generation
CN102633880B (zh) 2003-05-02 2015-02-25 赞科股份有限公司 优化的Fc变体及其产生方法
RU2337107C2 (ru) 2003-05-02 2008-10-27 Ксенкор, Инк. ОПТИМИЗИРОВАННЫЕ Fc-ВАРИАНТЫ, ИМЕЮЩИЕ ИЗМЕНЕННОЕ СВЯЗЫВАНИЕ С FcγR, И СПОСОБЫ ИХ ПОЛУЧЕНИЯ
US20050095243A1 (en) 2003-06-05 2005-05-05 Genentech, Inc. Combination therapy for B cell disorders
US20070269371A1 (en) 2003-09-05 2007-11-22 Genentech, Inc. Antibodies with altered effector functions
JP2005101105A (ja) 2003-09-22 2005-04-14 Canon Inc 位置決め装置、露光装置、デバイス製造方法
US20070041978A1 (en) 2003-10-10 2007-02-22 Kunihiro Hattori Bispecific antibody substituting for functional proteins
WO2005035756A1 (ja) 2003-10-10 2005-04-21 Chugai Seiyaku Kabushiki Kaisha 機能蛋白質を代替する二種特異性抗体
WO2005037867A1 (en) 2003-10-15 2005-04-28 Pdl Biopharma, Inc. ALTERATION OF Fc-FUSION PROTEIN SERUM HALF-LIVES BY MUTAGENESIS OF POSITIONS 250, 314 AND/OR 428 OF THE HEAVY CHAIN CONSTANT REGION OF IG
US20070037734A1 (en) 2003-11-05 2007-02-15 Ares Trading S.A. Process for the purification of il-18 binding protein
WO2005047327A2 (en) 2003-11-12 2005-05-26 Biogen Idec Ma Inc. NEONATAL Fc RECEPTOR (FcRn)-BINDING POLYPEPTIDE VARIANTS, DIMERIC Fc BINDING PROTEINS AND METHODS RELATED THERETO
US20070148164A1 (en) * 2003-11-12 2007-06-28 Biogen Idec Ma Inc. Neonatal Fc receptor (FcRn)-binding polypeptide variants, dimeric Fc binding proteins and methods related thereto
WO2005059106A2 (en) 2003-12-10 2005-06-30 Medarex, Inc. Interferon alpha antibodies and their uses
WO2005077981A2 (en) 2003-12-22 2005-08-25 Xencor, Inc. Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES
WO2005067620A2 (en) 2004-01-09 2005-07-28 Pfizer Inc. ANTIBODIES TO MAdCAM
US20080089892A1 (en) * 2004-01-12 2008-04-17 Eli Lilly And Co. Fc Region Variants
WO2005070963A1 (en) 2004-01-12 2005-08-04 Applied Molecular Evolution, Inc Fc region variants
US20050244403A1 (en) 2004-03-24 2005-11-03 Xencor, Inc. Immunoglobulin variants outside the Fc region
US20050260711A1 (en) 2004-03-30 2005-11-24 Deepshikha Datta Modulating pH-sensitive binding using non-natural amino acids
WO2005123780A2 (en) 2004-04-09 2005-12-29 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
WO2005112564A2 (en) 2004-04-15 2005-12-01 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Germline and sequence variants of humanized antibodies and methods of making and using them
JP2007532139A (ja) 2004-04-16 2007-11-15 マクロジェニクス,インコーポレーテッド FcγRIIB特異的抗体とその利用法
US20050260213A1 (en) 2004-04-16 2005-11-24 Scott Koenig Fcgamma-RIIB-specific antibodies and methods of use thereof
WO2005115452A2 (en) 2004-04-16 2005-12-08 Macrogenics, Inc. Fcϝriib-specific antibodies and methods of use thereof
WO2005123126A2 (en) 2004-06-09 2005-12-29 Wyeth Antibodies against human interleukin-13 and uses therefor
US20060063228A1 (en) 2004-06-09 2006-03-23 Kasaian Marion T Antibodies against human interleukin-13 and uses therefor
US20060153860A1 (en) 2004-06-18 2006-07-13 Ambrx, Inc. Novel antigen-binding polypeptides and their uses
WO2006004663A2 (en) 2004-06-25 2006-01-12 Medimmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
EP1773391A2 (en) 2004-06-25 2007-04-18 MedImmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
US20060019342A1 (en) 2004-06-25 2006-01-26 Medimmune, Inc. Increasing the production of recombinant antibodies in mammalian cells by site-directed mutagenesis
WO2006019447A1 (en) 2004-07-15 2006-02-23 Xencor, Inc. Optimized fc variants
CN101014619A (zh) 2004-07-15 2007-08-08 赞科股份有限公司 优化的Fc变体
JP2008505174A (ja) 2004-07-15 2008-02-21 ゼンコー・インコーポレイテッド 最適化Fc変異体
CN101001873A (zh) 2004-08-04 2007-07-18 应用分子进化公司 Fc区变体
WO2006020114A2 (en) 2004-08-04 2006-02-23 Applied Molecular Evolution, Inc. Variant fc regions
JP2008511292A (ja) 2004-08-04 2008-04-17 アプライド モレキュラー エボリューション,インコーポレイテッド 変異Fc領域
WO2006016644A1 (ja) 2004-08-11 2006-02-16 Mitsubishi Chemical Corporation 抗体及びその利用
EP1787998A1 (en) 2004-08-11 2007-05-23 Mitsubishi Chemical Corporation Antibody and utilization of the same
WO2006023403A2 (en) 2004-08-16 2006-03-02 Medimmune, Inc. Eph receptor fc variants with enhanced antibody dependent cell-mediated cytotoxicity activity
RU2367667C2 (ru) 2004-08-19 2009-09-20 Дженентек, Инк. Полипептидные варианты с измененной эффекторной функцией
JP2008510466A (ja) 2004-08-19 2008-04-10 ジェネンテック・インコーポレーテッド エフェクター機能が変更しているポリペプチド変異体
US20060067930A1 (en) 2004-08-19 2006-03-30 Genentech, Inc. Polypeptide variants with altered effector function
WO2006031370A2 (en) 2004-08-19 2006-03-23 Genentech, Inc. Polypeptide variants with altered effector function
WO2006030200A1 (en) 2004-09-14 2006-03-23 National Institute For Biological Standards And Control Vaccine
WO2006030220A1 (en) 2004-09-17 2006-03-23 Domantis Limited Compositions monovalent for cd40l binding and methods of use
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US20090053240A1 (en) 2004-10-21 2009-02-26 Xencor, Inc. Novel Immunoglobulin Insertions, Deletions and Substitutions
WO2006050166A2 (en) 2004-10-29 2006-05-11 Medimmune, Inc. Methods of preventing and treating rsv infections and related conditions
WO2006050491A2 (en) 2004-11-08 2006-05-11 Epitomics, Inc. Methods for antibody engineering
US20060134709A1 (en) 2004-11-10 2006-06-22 Jeffery Stavenhagen Engineering Fc antibody regions to confer effector function
WO2007024249A2 (en) 2004-11-10 2007-03-01 Macrogenics, Inc. Engineering fc antibody regions to confer effector function
WO2006053301A2 (en) 2004-11-12 2006-05-18 Xencor, Inc. Fc variants with altered binding to fcrn
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
RU2007121679A (ru) 2004-11-12 2008-12-20 Ксенкор Fc-ВАРИАНТЫ С ИЗМЕНЕННЫМ СВЯЗЫВАНИЕМ С FcRn
CN101098890A (zh) 2004-11-12 2008-01-02 赞科股份有限公司 对FcRn的结合被改变的Fc变体
EP2314618A2 (en) 2004-11-12 2011-04-27 Xencor Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US20090041770A1 (en) 2004-11-12 2009-02-12 Chamberlain Aaron Keith Fc VARIANTS WITH ALTERED BINDING TO FcRn
JP2008519860A (ja) 2004-11-12 2008-06-12 ゼンコー・インコーポレイテッド FcRnとの変化した結合を有するFc変異体
US20080166756A1 (en) 2004-12-22 2008-07-10 Masayuki Tsuchiya Method of Producing an Antibody Using a Cell in Which the Function of Fucose Transporter Is Inhibited
WO2006067913A1 (ja) 2004-12-22 2006-06-29 Chugai Seiyaku Kabushiki Kaisha フコーストランスポーターの機能が阻害された細胞を用いた抗体の作製方法
WO2006066598A2 (en) 2004-12-23 2006-06-29 Novo Nordisk A/S Antibody binding affinity ligands
EP1870459A1 (en) 2005-03-31 2007-12-26 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
WO2006105338A2 (en) 2005-03-31 2006-10-05 Xencor, Inc. Fc VARIANTS WITH OPTIMIZED PROPERTIES
WO2006106905A1 (ja) 2005-03-31 2006-10-12 Chugai Seiyaku Kabushiki Kaisha 会合制御によるポリペプチド製造方法
US20100003254A1 (en) 2005-04-08 2010-01-07 Chugai Seiyaku Kabushiki Kaisha Antibody Substituting for Function of Blood Coagulation Factor VIII
WO2006109592A1 (ja) 2005-04-08 2006-10-19 Chugai Seiyaku Kabushiki Kaisha 血液凝固第viii因子の機能代替抗体
WO2006116260A2 (en) 2005-04-26 2006-11-02 Medimmune, Inc. Modulation of antibody effector function by hinge domain engineering
WO2006121852A2 (en) 2005-05-05 2006-11-16 Duke University Anti-cd19 antibody therapy for autoimmune disease
WO2006130834A2 (en) 2005-05-31 2006-12-07 Board Of Regents, The University Of Texas System IGGl ANTIBODIES WITH MUTATED FC PORTION FOR INCREASED BINDING TO FCRN RECEPTOR AND USES THEREOF
WO2007021841A2 (en) 2005-08-10 2007-02-22 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
WO2007024535A2 (en) 2005-08-19 2007-03-01 Wyeth Antagonist antibodies against gdf-8 and uses in treatment of als and other gdf-8-associated disorders
WO2007041635A2 (en) 2005-10-03 2007-04-12 Xencor, Inc. Fc variants with optimized fc receptor binding properties
US20070160598A1 (en) 2005-11-07 2007-07-12 Dennis Mark S Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
WO2007060411A1 (en) 2005-11-24 2007-05-31 Ucb Pharma S.A. Anti-tnf alpha antibodies which selectively inhibit tnf alpha signalling through the p55r
US20080292637A1 (en) 2005-11-30 2008-11-27 Pnina Fishman Therapeutic Uses of A3 Adenosine Receptor Antibodies
WO2007076524A2 (en) 2005-12-29 2007-07-05 Centocor, Inc. Human anti-il-23 antibodies, compositions, methods and uses
EP2009101A1 (en) 2006-03-31 2008-12-31 Chugai Seiyaku Kabushiki Kaisha Antibody modification method for purifying bispecific antibody
EP2006381A1 (en) 2006-03-31 2008-12-24 Chugai Seiyaku Kabushiki Kaisha Method for control of blood kinetics of antibody
CA2647846A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
WO2007114319A1 (ja) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha 抗体の血中動態を制御する方法
JP2013165716A (ja) 2006-03-31 2013-08-29 Chugai Pharmaceut Co Ltd 抗体の血中動態を制御する方法
WO2007114325A1 (ja) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha 二重特異性抗体を精製するための抗体改変方法
US20090324589A1 (en) 2006-03-31 2009-12-31 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
JP5144499B2 (ja) 2006-03-31 2013-02-13 中外製薬株式会社 二重特異性抗体を精製するための抗体改変方法
US20090263392A1 (en) 2006-03-31 2009-10-22 Chugai Seiyaku Kabushiki Kaisha Methods of modifying antibodies for purification of bispecific antibodies
US20070253951A1 (en) 2006-04-24 2007-11-01 Gordon Ng Humanized c-Kit antibody
US20080044417A1 (en) 2006-05-26 2008-02-21 Macrogenics, Inc. Humanized Fc.gamma.RIIB-Specific Antibodies and Methods of Use Thereof
WO2007142325A1 (ja) 2006-06-08 2007-12-13 Chugai Seiyaku Kabushiki Kaisha 炎症性疾患の予防または治療剤
WO2008002933A2 (en) 2006-06-26 2008-01-03 Macrogenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
WO2008022152A2 (en) 2006-08-14 2008-02-21 Xencor, Inc. Optimized antibodies that target cd19
WO2008060785A2 (en) 2006-10-06 2008-05-22 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Prevention of tissue ischemia, related methods and compositions
US20100216187A1 (en) 2006-10-11 2010-08-26 Ablynx N.V. Amino acid sequences that bind to a desired molecule in a conditional manner
JP2010505436A (ja) 2006-10-11 2010-02-25 アブリンクス エン.ヴェー. コンディショナルな方法で所望の分子に結合するアミノ酸配列
WO2008043822A2 (en) 2006-10-11 2008-04-17 Ablynx N. V. Amino acid sequences that bind to a desired molecule in a conditional manner
US20080138349A1 (en) 2006-12-08 2008-06-12 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2008091954A2 (en) 2007-01-23 2008-07-31 Xencor, Inc. Optimized cd40 antibodies and methods of using the same
JP2010514460A (ja) 2007-01-23 2010-05-06 ゼンコー・インコーポレイテッド 最適化cd40抗体および前記を使用する方法
WO2008092117A2 (en) 2007-01-25 2008-07-31 Xencor, Inc. Immunoglobulins with modifications in the fcr binding region
US20100184959A1 (en) 2007-03-19 2010-07-22 Medimmune Limited Polypeptide Variants
WO2008143954A2 (en) 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Single-chain fc (scfc) regions, binding polypeptides comprising same, and methods related thereto
US20090136485A1 (en) 2007-05-30 2009-05-28 Xencor, Inc. Methods and compositions for inhibiting CD32B expressing cells
US20120321620A1 (en) 2007-05-30 2012-12-20 Xencor, Inc. Methods and compositions for inhibiting cd32b expressing cells
WO2009006338A1 (en) 2007-06-29 2009-01-08 Quest Diagnostics Investments Incorporated Analysis of amino acids in body fluid by liquid chromatography-mass spectrometry
EP2189526A1 (en) 2007-07-06 2010-05-26 Tokyo Metropolitan Organization for Medical Research Antibody binding specifically to tdp-43 aggregate
WO2009008529A1 (ja) 2007-07-06 2009-01-15 Tokyo Metropolitan Organization For Medical Research Tdp-43凝集物に特異的に結合する抗体
EP2202245A1 (en) 2007-09-26 2010-06-30 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
US20110076275A1 (en) 2007-09-26 2011-03-31 Chugai Seiyaku Kabushiki Kaisha Method of Modifying Isoelectric Point of Antibody Via Amino Acid Substitution in CDR
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US20110245473A1 (en) 2007-09-26 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Anti-IL-6 Receptor Antibody
RU2010116152A (ru) 2007-09-26 2011-11-10 Чугаи Сейяку Кабусики Кайся (Jp) Антитело против рецептора il-6
JP5334319B2 (ja) 2007-09-26 2013-11-06 中外製薬株式会社 Cdrのアミノ酸置換により抗体の等電点を改変する方法
JP2015130883A (ja) 2007-09-26 2015-07-23 中外製薬株式会社 Cdrのアミノ酸置換により抗体の等電点を改変する方法
US20180142027A1 (en) 2007-09-26 2018-05-24 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
US9096651B2 (en) 2007-09-26 2015-08-04 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US20100298542A1 (en) 2007-09-26 2010-11-25 Chugai Seiyaku Kabushiki Kaisha Modified Antibody Constant Region
WO2009041643A1 (ja) 2007-09-26 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Cdrのアミノ酸置換により抗体の等電点を改変する方法
US20150284465A1 (en) 2007-09-26 2015-10-08 Chugai Seiyaku Kabushiki Kaisha Method of Modifying Isoelectric Point of Antibody Via Amino Acid Substitution in CDR
EP2196541A1 (en) 2007-09-28 2010-06-16 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
US20150315278A1 (en) 2007-09-28 2015-11-05 Chugai Seiyaku Kabushiki Kaisha Anti-Glypican-3 Antibody Having Improved Kinetics in Plasma
US20100239577A1 (en) 2007-09-28 2010-09-23 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
WO2009041062A1 (ja) 2007-09-28 2009-04-02 Chugai Seiyaku Kabushiki Kaisha 血漿中動態が改善されたグリピカン3抗体
CA2700986A1 (en) 2007-09-28 2009-04-02 Chugai Seiyaku Kabushiki Kaisha Anti-glypican-3 antibody having improved kinetics in plasma
WO2009053358A1 (en) 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying fc-fusion proteins
WO2009058492A2 (en) 2007-10-31 2009-05-07 Xencor, Inc Fc variants with altered binding to fcrn
WO2009062083A2 (en) 2007-11-08 2009-05-14 Pikamab, Inc. Methods and compositions for antibody therapy
US20110044986A1 (en) 2007-12-21 2011-02-24 Amgen Inc. Anti-amyloid antibodies and uses thereof
WO2009086320A1 (en) 2007-12-26 2009-07-09 Xencor, Inc Fc variants with altered binding to fcrn
WO2009095235A1 (en) 2008-01-29 2009-08-06 Ablynx N.V. Methods to stabilize proteins and polypeptides
US20130011866A1 (en) 2008-04-11 2013-01-10 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
WO2009125825A1 (ja) 2008-04-11 2009-10-15 中外製薬株式会社 複数分子の抗原に繰り返し結合する抗原結合分子
US9868948B2 (en) 2008-04-11 2018-01-16 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
TW201000127A (en) 2008-04-11 2010-01-01 Chugai Pharmaceutical Co Ltd Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US20130303396A1 (en) 2008-04-11 2013-11-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
JPWO2009125825A1 (ja) * 2008-04-11 2011-08-04 中外製薬株式会社 複数分子の抗原に繰り返し結合する抗原結合分子
KR20110004435A (ko) 2008-04-11 2011-01-13 추가이 세이야쿠 가부시키가이샤 복수 분자의 항원에 반복 결합하는 항원 결합 분자
CN102056946A (zh) 2008-04-11 2011-05-11 中外制药株式会社 与多个分子的抗原反复结合的抗原结合分子
EP2275443A1 (en) 2008-04-11 2011-01-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US9890377B2 (en) 2008-04-11 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US20130336963A1 (en) 2008-04-11 2013-12-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US20110111406A1 (en) * 2008-04-11 2011-05-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
WO2009139822A1 (en) 2008-05-01 2009-11-19 Amgen Inc. Anti-hepcidin antibodies and methods of use
US20110150888A1 (en) 2008-05-01 2011-06-23 Amgen Inc. Anti-hepcidin antibodies and methods of use
WO2009155513A2 (en) 2008-06-20 2009-12-23 Novartis Ag Immunoglobulins with reduced aggregation
US8562991B2 (en) 2008-09-26 2013-10-22 Chugai Seiyaku Kabushiki Kaisha Antibody molecules that bind to IL-6 receptor
JP2010079667A (ja) 2008-09-26 2010-04-08 Kyocera Mita Corp 画像処理装置、画像処理方法及び画像処理プログラム
US20110098450A1 (en) 2008-09-26 2011-04-28 Chugai Seiyaku Kabushiki Kaisha Antibody Molecules
RU2430111C1 (ru) 2008-09-26 2011-09-27 Чугаи Сейяку Кабусики Кайся Молекулы антител с улучшенными свойствами
JP2012505833A (ja) 2008-10-14 2012-03-08 ジェネンテック, インコーポレイテッド 免疫グロブリン変異体及びその用途
US20100098730A1 (en) 2008-10-14 2010-04-22 Lowman Henry B Immunoglobulin variants and uses thereof
WO2010045193A1 (en) 2008-10-14 2010-04-22 Genentech, Inc. Immunoglobulin variants and uses thereof
WO2010058860A1 (ja) 2008-11-18 2010-05-27 株式会社シノテスト 試料中のc反応性蛋白質の測定方法及び測定試薬
WO2010081173A2 (en) 2009-01-12 2010-07-15 Cytomx Therapeutics, Llc Modified antibody compositions, methods of making and using thereof
WO2010085682A2 (en) 2009-01-23 2010-07-29 Biogen Idec Ma Inc. Stabilized fc polypeptides with reduced effector function and methods of use
US20100292443A1 (en) 2009-02-26 2010-11-18 Sabbadini Roger A Humanized platelet activating factor antibody design using anti-lipid antibody templates
WO2010107109A1 (ja) 2009-03-19 2010-09-23 中外製薬株式会社 抗体定常領域改変体
WO2010106180A2 (en) 2009-03-20 2010-09-23 Lfb Biotechnologies Optimized fc variants
JP2010250830A (ja) 2009-04-16 2010-11-04 Accenture Global Services Gmbh ウェブ・サイト・アクセラレータ
US8568726B2 (en) * 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
WO2011043643A1 (en) 2009-10-06 2011-04-14 Medimmune Ltd Rsv-specific binding molecule
WO2011044368A1 (en) 2009-10-07 2011-04-14 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2011094593A2 (en) 2010-01-28 2011-08-04 Ab Biosciences, Inc. Novel lowered affinity antibodies and methods of marking the same
JP2013518131A (ja) 2010-01-28 2013-05-20 アブ バイオサイエンシズ インコーポレイテッド 親和性が低下した抗体およびそれを作製する方法
US8329867B2 (en) 2010-02-19 2012-12-11 Xencor, Inc. CTLA4-Ig immunoadhesins
SG183867A1 (en) 2010-03-11 2012-10-30 Rinat Neuroscience Corp ANTIBODIES WITH pH DEPENDENT ANTIGEN BINDING
US9029515B2 (en) 2010-03-11 2015-05-12 Rinat Neuroscience Corp. Anti-PCSK9 antibodies with pH dependent antigen binding
US20110229489A1 (en) 2010-03-11 2011-09-22 Rinat Neuroscience Corp. Antibodies with pH Dependent Antigen Binding
JP2011184418A (ja) 2010-03-11 2011-09-22 Tokyo Institute Of Technology 親和性可変抗体
WO2011111007A2 (en) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation ANTIBODIES WITH pH DEPENDENT ANTIGEN BINDING
US20130131319A1 (en) 2010-03-30 2013-05-23 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
US20170226206A1 (en) 2010-03-30 2017-08-10 Chugai Seiyaku Kabushiki Kaisha Antibodies With Modified Affinity To FcRn That Promote Antigen Clearance
US20160244526A1 (en) 2010-03-30 2016-08-25 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
TW201202419A (en) 2010-03-30 2012-01-16 Chugai Pharmaceutical Co Ltd Antigen-binding molecules that promote antigen clearance
WO2011122011A2 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
CA2794860A1 (en) 2010-03-30 2011-10-06 Chugai Seiyaku Kabushiki Kaisha Antibodies with modified affinity to fcrn that promote antigen clearance
WO2012033953A1 (en) 2010-09-08 2012-03-15 Halozyme, Inc. Methods for assessing and identifying or evolving conditionally active therapeutic proteins
US20180258161A1 (en) 2010-11-30 2018-09-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
WO2012073992A1 (ja) 2010-11-30 2012-06-07 中外製薬株式会社 複数分子の抗原に繰り返し結合する抗原結合分子
US20140234340A1 (en) 2010-11-30 2014-08-21 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
EP2647706A1 (en) 2010-11-30 2013-10-09 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US20140105889A1 (en) 2011-02-25 2014-04-17 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
CN103492565A (zh) 2011-02-25 2014-01-01 中外制药株式会社 FcγRIIb特异性Fc抗体
EP2679681A1 (en) 2011-02-25 2014-01-01 Chugai Seiyaku Kabushiki Kaisha Fc riib-specific fc antibody
US20140093496A1 (en) 2011-02-25 2014-04-03 Chugai Seiyaku Kabushiki Kaisha Fc-gamma-RIIb-SPECIFIC Fc ANTIBODY
CA2827923A1 (en) 2011-02-25 2012-08-30 Chugai Seiyaku Kabushiki Kaisha Fc.gamma.riib-specific fc antibody
WO2012115241A1 (ja) 2011-02-25 2012-08-30 中外製薬株式会社 FcγRIIb特異的Fc抗体
SG192945A1 (en) 2011-02-25 2013-09-30 Chugai Pharmaceutical Co Ltd Fcgriib-specific fc antibody
US20140112926A1 (en) 2011-03-16 2014-04-24 Amgen Inc. Fc VARIANTS
WO2012132067A1 (ja) 2011-03-30 2012-10-04 中外製薬株式会社 抗原結合分子の血漿中滞留性と免疫原性を改変する方法
CA2831770A1 (en) 2011-03-30 2012-10-04 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
WO2012133782A1 (ja) 2011-03-30 2012-10-04 中外製薬株式会社 抗原結合分子の血漿中滞留性と免疫原性を改変する方法
EP2698431A1 (en) 2011-03-30 2014-02-19 Chugai Seiyaku Kabushiki Kaisha Retention of antigen-binding molecules in blood plasma and method for modifying immunogenicity
US20140086916A1 (en) 2011-05-25 2014-03-27 Dongxing Zha METHOD FOR PREPARING Fc CONTAINING POLYPEPTIDES HAVING IMPROVED PROPERTIES
WO2013004842A2 (en) 2011-07-06 2013-01-10 Genmab A/S Antibody variants and uses thereof
WO2013047752A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 抗原の消失を促進する抗原結合分子
US20140255398A1 (en) 2011-09-30 2014-09-11 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule inducing immune response to target antigen
US20140335089A1 (en) 2011-09-30 2014-11-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting elimination of antigens
US20170022270A1 (en) 2011-09-30 2017-01-26 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
WO2013047748A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 複数の生理活性を有する抗原の消失を促進する抗原結合分子
EP2762166A1 (en) 2011-09-30 2014-08-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting loss of antigens
US10024867B2 (en) * 2011-09-30 2018-07-17 Chugai Seiyaku Kabushiki Kaisha Ion concentration-dependent binding molecule library
WO2013047729A1 (ja) 2011-09-30 2013-04-04 中外製薬株式会社 標的抗原に対する免疫応答を誘導する抗原結合分子
US20150050269A1 (en) 2011-09-30 2015-02-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule promoting disappearance of antigens having plurality of biological activities
US20150299313A1 (en) 2011-10-05 2015-10-22 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for promoting clearance from plasma of antigen comprising suger chain receptor-binding domain
WO2013081143A1 (ja) 2011-11-30 2013-06-06 中外製薬株式会社 免疫複合体を形成する細胞内への運搬体(キャリア)を含む医薬
US20150056182A1 (en) 2011-11-30 2015-02-26 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US20150210763A1 (en) 2012-02-09 2015-07-30 Chugai Seiyaku Kabushiki Kaisha MODIFIED Fc REGION OF ANTIBODY
WO2013125667A1 (ja) 2012-02-24 2013-08-29 中外製薬株式会社 FcγRIIBを介して抗原の消失を促進する抗原結合分子
US20160046693A1 (en) 2012-02-24 2016-02-18 Chugai Seiyaku Kabushiki Kaisha Antigen-Binding Molecule for Promoting Disappearance of Antigen via Fc gamma RIIB
WO2013180201A1 (ja) 2012-05-30 2013-12-05 中外製薬株式会社 会合化した抗原を消失させる抗原結合分子
WO2013180200A1 (ja) 2012-05-30 2013-12-05 中外製薬株式会社 標的組織特異的抗原結合分子
US20150166654A1 (en) 2012-05-30 2015-06-18 Chugai Seiyaku Kabushiki Kaisha Target tissue-specific antigen-binding molecule
US20150353630A1 (en) 2012-05-30 2015-12-10 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule for eliminating aggregated antigens
WO2014028354A1 (en) 2012-08-13 2014-02-20 Regeneron Pharmaceuticals, Inc. Anti-pcsk9 antibodies with ph-dependent binding characteristics
US20150203577A1 (en) 2012-08-24 2015-07-23 Chugai Seiyaku Kabushiki Kaisha MOUSE FcgammaRII-SPECIFIC Fc ANTIBODY
WO2014030728A1 (ja) 2012-08-24 2014-02-27 中外製薬株式会社 FcγRIIb特異的Fc領域改変体
EP2889377A1 (en) 2012-08-24 2015-07-01 Chugai Seiyaku Kabushiki Kaisha Fc riib-specific fc region variant
US20150299296A1 (en) 2012-08-24 2015-10-22 Chugai Seiyaku Kabushiki Kaisha FCgammaRIIB-SPECIFIC FC REGION VARIANT
US20150247849A1 (en) 2012-09-21 2015-09-03 Paul P. Tamburini Screening assays for complement component c5 antagonists
US20160068592A1 (en) 2013-01-31 2016-03-10 Seoul National University R&Db Foundation C5 antibody and method for preventing and treating complement-related diseases
WO2014144575A1 (en) 2013-03-15 2014-09-18 Forta Corporation Reinforcement fiber coating compositions, methods of making and treating, and uses for improved adhesion to asphalt and portland cement concrete
WO2014144080A2 (en) 2013-03-15 2014-09-18 Amgen Inc. Human antigen binding proteins that bind to proprotein convertase subtilisin kexin type 9
US20160039912A1 (en) 2013-04-02 2016-02-11 Chugai Seiyaku Kabushiki Kaisha Fc REGION VARIANT
WO2014163101A1 (ja) 2013-04-02 2014-10-09 中外製薬株式会社 Fc領域改変体
US9079949B1 (en) 2014-03-07 2015-07-14 Alexion Pharmaceuticals, Inc. Anti-C5 antibodies having improved pharmacokinetics
WO2016000813A1 (en) 2014-06-30 2016-01-07 Merck Patent Gmbh Anti-tnfa antibodies with ph-dependent antigen binding
US9765135B2 (en) * 2014-12-19 2017-09-19 Chugai Seiyaku Kabushiki Kaisha Anti-C5 antibodies
US9969800B2 (en) * 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies

Non-Patent Citations (540)

* Cited by examiner, † Cited by third party
Title
Adams et al., "Humanization of a recombinant monoclonal antibody to produce a therapeutic HER dimerization inhibitor, pertuzumab," Cancer Immunol. Immunother., 55:717-727 (2006).
Akbarzadeh-Sharbaf et al., "In silico design, construction and cloning of Trastuzumab humanized monoclonal antibody: A possible biosimilar for Herceptin," Adv Biomed Res, Jan.-Mar. 2012, 1(1):1-6. doi: 10. 4103/ 2277-9175. 98122. Epub Jul. 6, 2012.
Algonomics-TripoleR applications [Online], Retrieved from the Internet on Feb. 29, 2012: http://www.algonomics.com/proteinengineering/tripole_applications.php, 2 pages, available online on Feb. 21, 2009.
Algonomics—TripoleR applications [Online], Retrieved from the Internet on Feb. 29, 2012: http://www.algonomics.com/proteinengineering/tripole_applications.php, 2 pages, available online on Feb. 21, 2009.
Alignment of constant region sequences from WO 2009/125825 (document submitted in EP opposition and posted by EPO on Feb. 2, 2018); 1 page.
Alignment of the amino acid sequences of the Fc regions of antibodies exemplified in EP 2275443 (document submitted in EP opposition and posted by EPO on Feb. 2, 2018); 1 page.
Alignment of variable heavy and light chain amino acid sequences from WO 2009/125825 (document submitted in EP opposition and posted by EPO on Feb. 2, 2018); 2 pages.
Alley et al., "Antibody-drug conjugates: targeted drug delivery for cancer," Curr Opin Chem Biol., Aug. 2010; 14(4):529-37. doi: 10.1016/j.cbpa.2010.06.170. Epub Jul. 17, 2010.
Almagro et al., "Humanization of antibodies," Front. Biosci. Jan. 1, 2008; 13:1619-33.
Amersham Biosciences, "Affinity Chromatography: Principles and Methods," Edition AD, pp. 16-18, 137 (2002).
Amersham Biosciences, "Antibody Purification Handbook," Edition 18-1037-46 [online], [retrieved on Nov. 5, 2015]. Retrieved from the Internet: http://www.promix.ru/manuf/ge/chrom/lit/Antibody_Purification.pdf.
Amigorena et al., "Cytoplasmic domain heterogeneity and functions of IgG Fc receptors in B lymphocytes," Science, 256(5065):1808-12 (1992).
Amigorena et al., "Fc gamma RII expression in resting and activated B lymphocytes," Eur J Immunol., (8):1379-85 (1989).
Antibodies from www.bioinf.org.uk: Dr. Andrew C.R. Martin's Group, downloaded Jul. 11, 2018, nine pages.
Araujo et al., "Increased rheumatoid factor interference observed during immunogenicity assessment of an Fe-engineered therapeutic antibody," J. Pharm. Biomed. Anlys. Jul. 15, 2011;55(5):1041-9. doi: 10.1016/j.jpba.2011.03.008. Epub Mar. 11, 2011.
Armour et al., "Differential binding to human FcgammaRIIa and FcgammaRIIb receptors by human IgG wildtype and mutant antibodies," Mol Immunol., 40(9):585-93 (2003).
Armour et al., "Recombinant human IgG molecules lacking Fcγ receptor I binding and monocyte triggering activities," Eur. J. Immunol. Aug. 5, 1999; 29(8):2613-24.
Atherton et al., "Acid-base balance: maintenance of plasma pH," Anaesthesia & Intensive Care Medicine, 2009, 10(11):557-61 (abstract).
Baeuerle et al., "BiTE: Teaching antibodies to engage T-cells for cancer therapy," Curr Opin Mol Ther., 11(1):22-30 (2009).
Balint et al., "Antibody engineering by parsimonious mutagenesis," Gene., Dec. 27, 1993;137(1):109-18.
Bartelds et al., "Clinical response to adalimumab: relationship to anti-adalimumab antibodies and serum adalimumab concentrations in rheumatoid arthritis," Ann Rheum. Dis., 66:921-926 (2007).
Batra et al., "Pharmacokinetics and biodistribution of genetically engineered antibodies," Curr Opin Biotechnol., Dec. 2002;13(6):603-8.
Bayry et al., "Immuno affinity purification of foot and mouth disease virus type specific antibodies using recombinant protein absorbed to polystyrene wells," J. Virol. Methods. Aug. 1999; 81(1-2):21-30.
Beck et al., "Strategies and challenges for the next generation of therapeutic antibodies," Nat Rev Immunol., 10(5):345-52 (2010).
Bender et al., "Immunogenicity, efficacy and adverse events of adalimumab in RA patients," Rheumatol. Int., 27:269-274 (2007).
Beringhelli et al., "pH and ionic strength dependence of protein (un)folding and ligand binding to bovine beta-lactoglobulins A and B," Biochemistry, Dec. 24, 2002;41(51):15415-22.
Biasini et al., "Immunopurification of pathological prion protein aggregates," PLoS One, Nov. 12, 2009;4(11):e7816. doi: 10.1371/journal.pone.0007816.
Binz et al., "Engineering novel binding proteins from nonimmunoglobulin domains," Nat. Biotechnol., 23:1257-68 (2005).
Blank et al., Decreased transcription of the human FCGR2B gene mediated by the -343 G/C promoter polymorphism and association with systemic lupus erythematosus. Hum Genet., 117(2-3):220-7 (2005).
Boruchov et al., "Activating and inhibitory IgG Fc receptors on human DCs mediate opposing functions," J Clin Invest., 115(10):2914-23 (2005).
Boumpas et al., "A short course of BG9588 (anti-CD40 ligand antibody) improves serologic activity and decreases hematuria in patients with proliferative lupus glomerulonephritis," Arthritis Rheum., 48(3):719-27 (2003).
Brown et al., "A study of the interactions between an IgG-binding domain based on the B domain of staphylococcal protein A and rabbit IgG," Mol Biotechnol., Aug. 1998;10(1):9-16.
Brown et al., "Tolerance of single, but not multiple, amino acid replacements in antibody VH CDR2: a means of minimizing B cell wastage from somatic hypermutation?," J. Immunol., 156(9):3285-91 (1996).
Bruhns et al., Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses, Blood, 113(16):3716-25 (2009).
Bruhns, "Properties of mouse and human IgG receptors and their contribution to disease models," Blood, Jun. 14, 2012;119(24):5640-9. doi: 10.1182/blood-2012-01-380121. Epub Apr. 25, 2012.
Burmeister et al., "Crystal structure of the complex of rat neonatal Fc receptor with Fc," Nature, Nov. 24, 1994;372(6504):379-83.
Cartron et al., "Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene," Blood, 99(3):754-8 (2002).
Cemerski et al., "Suppression of mast cell degranulation through a dual-targeting tandem IgE-IgG Fc domain biologic engineered to bind with high affinity to FcγRIIb," Immunol Lett., 143(1):34-43 (2012).
Chaparro-Riggers et al., "Increasing serum half-life and extending cholesterol lowering in vivo by engineering antibody with pH-sensitive binding to PCSK9," J Biol Chem., 287(14):11090-7 (2012).
Chau et al., "HuM291(Nuvion), a humanized Fc receptor-nonbinding antibody against CD3, anergizes peripheral blood T cells as partial agonist of the T cell receptor," Transplantation Apr. 15, 2001; 71(7):941-50.
Chen et al., "Association of a transmembrane polymorphism of Fcgamma receptor IIb (FCGR2B) with systemic lupus erythematosus in Taiwanese patients," Arthritis Rheum., 54(12):3908-17 (2006).
Chen et al., "Defective Secretion of an Immunoglobulin Caused by Mutations in the Heavy Chain Complementarity Determining Region 2," Exp. Med. Aug. 1, 1994; 180(2):577-86.
Chen et al., "Enhancement and destruction of antibody function by somatic mutation: unequal occurrence is controlled by V gene combinatorial associations," EMBO J, Jun. 15, 1995, 14(12):2784-94.
Chen et al., "Generation and Analysis of Random Point Mutations in an Antibody CDR2 Sequence: Many Mutated Antibodies Lose Their Ability to Bind Antigen," J. Exp. Med. Sep. 1, 1992; 176(3):855-66.
Chen et al., "Selection and analysis of an optimized anti-VEGF antibody: crystal structure of an affinity-matured Fab in complex with antigen," J Mol Biol., Nov. 5, 1999;293(4):865-81.
Chirino et al., "Minimizing the immunogenicity of protein therapeutics," Drug Discov. Today., 9:82-90 (2004).
Chu et al., "Accumulation of Succinimide in a Recombinant Monoclonal Antibody in Mildly Acidic Buffers Under Elevated Temperatures," Pharm. Res. Jun. 2007; 24(6):1145-56. Epub Mar. 24, 2007.
Chu et al., "Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement of CD19 and FcgammaRIIb with Fc-engineered antibodies," Mol Immunol., 45(15):3926-33 (2008).
Chu et al., "Reduction of total IgE by targeted coengagement of IgE B-cell receptor and FcγRIIb with Fc-engineered antibody," J Allergy Clin Immunol., 129(4):1102-15 (2012).
Chuntharapai et al., "Isotype-dependent inhibition of tumor growth in vivo by monoclonal antibodies to death receptor 4," J Immunol., 166(8):4891-8 (2001).
Claims as granted for Publication No. EP 2275443, dated Jan. 19, 2011 (document submitted in EP opposition); 6 pages.
Clark, "An alignment of IgG sequences from Human, Mouse and Rat," Part II Immunoglobulin lectures (v4), pp. 5(i)-(ii) [retrieved on Jul. 25, 2014]. Retrieved from the Internet: http://www.path.cam.ac.uk/˜mrc7/lecturenotes/handout1a.pdf.
Clark, "IgG effector mechanisms," Chem Immunol., 65:88-110 (1997).
Clarkson et al., "Blockade of clearance of immune complexes by an anti-Fc gamma receptor monoclonal antibody," J Exp Med., Aug. 1, 1986;164(2):474-89.
Clynes et al., "Fc receptors are required in passive and active immunity to melanoma," Proc Natl Acad Sci USA, 95(2):652-6 (1998).
Clynes et al., "Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets," Nat Med., 6(4):443-6 (2000).
Cole et al., "Human IgC2 Variants of Chimeric Anti-CD3 Are Nonmitogenic to T Cells," J. Immunol. Oct. 1, 1997; 159(7):3613-21.
Colman et al., "Effects of amino acid sequence changes on antibody-antigen interactions," Res Immunol, Jan. 1994, 145(1):33-6.
Coloma et al., "Position effects of variable region carbohydrate on the affinity and in vivo behavior of an anti-(1-->6) dextran antibody," J Immunol., Feb. 15, 1999;162(4):2162-70.
Comper et al., "Charge selectivity in kidney ultrafiltration," Kidney Int. May 1995; 47(5):1242-51.
Cordoba et al., "Non-enzymatic hinge region fragmentation of antibodies in solution," J. Chromatogr. B-Analyt. Technol. Biomed. Life Sci. Apr. 25, 2005; 818(2):115-21.
Cordoba et al., "Non-enzymatic hinge region fragmentation of antibodies in solution," J. Chromatogr. B—Analyt. Technol. Biomed. Life Sci. Apr. 25, 2005; 818(2):115-21.
Couto et al., "Anti-BA46 Monoclonal Antibody Mc3: Humanization Using a Novel Positional Consensus and in Vivo and in Vitro Characterization," Cancer Res., 55:1717-22 (1995).
Cuatrecasas et al., "Affinity Chromatography", Methods Enzymol. 1971; 12:345-78.
Dall'Acqua et al., "Antibody humanization by framework shuffling," Methods, 36(1):43-60 (2005).
Dall'Acqua et al., "Increasing the affinity of a human IgG1 for the neonatal Fc receptor: biological consequences," J. Immunol., 169(9):5171-80 (2002).
Dall'Acqua et al., "Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn)," J Biol Chem., 281(33):23514-24 (2006).
Damschroder et al., "Framework shuffling of antibodies to reduce immunogenicity and manipulate functional and biophysical properties," Mol. Immunol. Apr. 2007; 44(11):3049-60. Epub Jan. 22, 2007.
Datta-Mannan et al., "Humanized IgG1 Variants with Differential Binding Properties to the Neonatal Fc Receptor: Relationship to Pharmacokinetics in Mice and Primates," Drug Metab Dispos, Jan. 2007, 35(1):86-94. Epub Oct. 18, 2006.
Datta-Mannan et al., "Monoclonal antibody clearance. Impact of modulating the interaction of IgG with the neonatal Fc receptor," J Biol Chem., Jan. 19, 2007;282(3):1709-17. Epub Nov. 29, 2006.
Davda et al., "Properties of a general PK/PD model of antibody-ligand interactions for therapeutic antibodies that bind to soluble endogenous targets," MAbs, Sep.-Oct.; 2(5):576-88. doi: 10.4161/mabs.2.5.12833. Epub Sep. 1, 2010.
Davydov, "Omalizuman (Xolair) for Treatment of Asthma," Am Fam Physician, Jan. 2005, 15:71(2):341-2.
De Bono et al., "ING-1, a monoclonal antibody targeting Ep-CAM in patients with advanced adenocarcinomas," Clin Cancer Res., 10(22):7555-65 (2004).
De Felice et al., "Formation of amyloid aggregates from human lysozyme and its disease-associated variants using hydrostatic pressure," FASEB J, Jul. 2004, 18(10):1099-101. (doi:10.1096/fj.03-1072fje; PMID 15155566).
De Groot et al., "De-immunization of Therapeutic Proteins by T-cell Epitope Modification," Dev. Biol. (Basel). Feb. 2005; 122:171-94.
Declaration of Nimish Gera, Ph.D., CV and Exhibits, dated Sep. 1, 2016, 24 pages.
Declaration of Nimish Gera, Ph.D., CV and Exhibits, Sep. 1, 2016 (submitted in the matter of EP 2275443, Opposition thereto by Alexion Pharmaceuticals, Inc.); 24 pages.
Deen et al., "Structural determinants of glomerular permeability," Am. J. Physiol. Renal Physiol. Oct. 1, 2001; 281(4): F579-96.
Del Rio et al., "An engineered penicillin acylase with altered surface charge is more stable in alkaline pH," Ann. N.Y. Acad. Sci. Oct. 12, 1996; 799:61-4.
Deng et al., "Pharmacokinetics of humanized monoclonal anti-tumor necrosis factor-{alpha} antibody and its neonatal Fc receptor variants in mice and cynomolgus monkeys," Drug Metab Dispos., Apr. 2010;38(4):600-5. doi: 10.1124/dmd.109.031310. Epub Jan. 13, 2010.
Desai et al., "Fc gamma receptor IIB on dendritic cells enforces peripheral tolerance by inhibiting effector T cell responses," J Immunol., 178(10):6217-26 (2007).
Desjarlais et al., "Optimizing engagement of the immune system by anti-tumor antibodies: an engineer's perspective," Drug Discov Today, Nov. 2007; 12(21-22):898-910. Epub Oct. 22, 2007.
Devanaboyina et al., "The effect of pH dependence of antibody-antigen interactions on subcellular trafficking dynamics," MAbs, 5(6):851-9 (2013).
Dhodapkar et al., "Selective blockade of inhibitory Fcgamma receptor enables human dendritic cell maturation with IL-12p70 production and immunity to antibody-coated tumor cells," Proc Natl Acad Sci USA, 102(8):2910-5 (2005).
Drake et al., "Chapter 5: Biophysical Considerations for Development of Antibody-Based Therapeutics," Biophysical Considerations for Development of Antibody-Based Therapeutics, Springer Springer Science+Business Media New York, 95-7 (2012).
Duffau et al., "Platelet CD154 potentiates interferon-alpha secretion by plasmacytoid dendritic cells in systemic lupus erythematosus," Sci Transl Med., 2(47):47ra63 (2010).
Dufner et al., "Harnessing phage and ribosome display for antibody optimization," Trends Biotechnol., Nov. 2006;24(11):523-9. Epub Sep. 26, 2006.
Durkee et al., "Immunoaffinity Chromatographic Purification of Russell's Viper Venom Factor X Activator Using Elution in High Concentrations of Magnesium Chloride," Protein Expr. Purif. Oct. 1993; 4(5):405-11.
Ejima et al., "Effective elution of antibodies by arginine and arginine derivatives in affinity column chromatography," Anal Biochem., Oct. 15, 2005;345(2):250-7.
EMA product information: Annexes to file of the tocilizumab preparation RoActemra (WC500054890), published Jan. 8, 2010, 109 pages.
EPO Register Extract EP 1915397 (document submitted in EP opposition and posted by EPO on Feb. 2, 2018); 4 pages.
Epstein, "Non-randomness of amino-acid changes in the evolution of homologous proteins," Nature, Jul. 22, 1967;215(5099):355-9.
Ewert et al., "Stability improvement of antibodies for extracellular and intracellular applications: CDR grafting to stable frameworks and structure-based framework engineering," Methods, 34:184-199 (2004).
Experimental data characterizing the binding of rituximab to its antigen CD20 and to human FcRn (document submitted in EP opposition and posted by EPO on Feb. 5, 2018); 6 pages.
Experimental information regarding off-rate of Xolair Fab for binding to human IgE at pH7.4 and pH5.5 (document submitted in EP opposition and posted by EPO on Feb. 2, 2018); 3 pages.
Expert Declaration by Dr. Madhusudan Natarajan, submitted in EP opposition regarding EP 2552955 and posted by EPO on Feb. 5, 2018; 4 pages.
Feinberg et al., "Mechanism of pH-dependent N-acetylgalactosamine binding by a functional mimic of the hepatocyte asialoglycoprotein receptor," J Biol Chem., 275(45):35176-84 (2000).
Fiedler et al., "An engineered IN-1 Fab fragment with improved affinity for the Nogo-A axonal growth inhibitor permits immunochemical detection and shows enhanced neutralizing activity," Protein Eng. Nov. 1, 2002: 15(11):931-41.
Fillipovic, Biochemical basis of human life activity. VLADOS, 2005:38-43 (with English translation).
Fillipovich, Biochemical basis of human life, VLADOS, 2005:49-50 (with English translation).
Finkelman et al., "Anti-cytokine antibodies as carrier proteins. Prolongation of in vivo effects of exogenous cytokines by injection of cytokine-anti-cytokine antibody complexes," J Immunol., 151(3):1235-44 (1993).
Fish & Richardson P.C., Amendment and Reply to Office Action dated Apr. 6, 2015 in U.S. Appl. No. 13/889,484, filed Jul. 6, 2015, 14 pages.
Fish & Richardson P.C., Amendment and Reply to Office Action dated Mar. 13, 2015 in U.S. Appl. No. 13/595,139, filed Jun. 11, 2015, 19 pages.
Fish & Richardson P.C., Amendment in Reply to Office Action dated Nov. 14, 2012 in U.S. Appl. No. 13/595,139, filed May 14, 2013, 19 pages.
Fish & Richardson P.C., Reply to Final Office Action dated Aug. 4, 2015 in U.S. Appl. No. 13/889,484, filed Dec. 2, 2015, 104 pages.
Fish & Richardson P.C., Reply to Final Office Action dated Oct. 16, 2015 in U.S. Appl. No. 14/347,034, filed Jan. 13, 2016, 28 pages.
Fish & Richardson P.C., Reply to Non-Final Office Action dated Apr. 16, 2015 in U.S. Appl. No. 14/347,034, filed Sep. 16, 2015, 28 pages.
Fish & Richardson P.C., Reply to Office Action dated Aug. 3, 2015 in U.S. Appl. No. 13/595,139, filed Dec. 2, 2015, 28 pages.
Fish & Richardson P.C., Reply to Office Action dated Feb. 12, 2016 in U.S. Appl. No. 13/595,139, filed Jul. 11, 2016, 4 pages.
Fish & Richardson P.C., Reply to Office Action dated Nov. 23, 2016 in U.S. Appl. No. 13/595,139, filed Mar. 22, 2016, 29 pages.
Fish & Richardson P.C., Reply to Restriction Requirement dated Dec. 17, 2015 in U.S. Appl. No. 14/347,321, filed Feb. 16, 2016, 3 pages.
Fish & Richardson P.C., Reply to Restriction Requirement dated Dec. 18, 2014 in U.S. Appl. No. 14/347,034, filed Mar. 18, 2015, 2 pages.
Fish & Richardson P.C., Response to Restriction Requirement dated Mar. 16, 2016 in U.S. Appl. No. 14/361,013, filed Aug. 1, 2016, 21 pages.
Fish & Richardson P.C., USPTO Response Non-Final Office Action in U.S. Appl. No. 14/361,013, dated Apr. 26, 2017, 117 pages.
Floto et al., "Loss of function of a lupus-associated FcgammaRIIb polymorphism through exclusion from lipid rafts," Nat Med., 11(10):1056-8 (2005).
Foote et al., "Antibody framework residues affecting the conformation of the hypervariable loops," J. Mol. Biol., Mar. 20, 1992; 224(2):487-99.
Fournier et al., "Activation of human peripheral IgM+ B cells is transiently inhibited by BCR-independent aggregation of Fc gammaRIIB," J Immunol., 181(8):5350-9 (2008).
Fujii, "Antibody affinity maturation by random mutagenesis," Antibody Engineering, Methods Mol. Biol. Series, 2004; 248:345-59.
GE Healthcare, Biacore, Sensor Surface Handbook BR-1005-71, Edition AB, Feb. 2005, pp. 1-100.
GE Healthcare. Application note 28-9277-92 AA. "High-throughput screening of elution pH for monoclonal antibodies on MabSelect SuRe using PreDictor plates" [online], [retrieved on Nov. 5, 2015]. Retrieved from the Internet: https://www.gelifesciences.com/gehcls_images/GELS/Related%20Content/Files/1314787424814/litdoc28927792AA_20110831131840.pdf.
Gera et al., "Design of pH Sensitive Binding Proteins from the Hyperthermophilic Sso7d Scaffold," PLoS One, 2012;7(11):e48928. doi: 10.1371/journal.pone.0048928. Epub Nov. 7, 2012.
Gerstner et al., "Sequence Plasticity in the Antigen-binding Site of a Therapeutic Anti-HER2 Antibody," J. Mol. Biol. Aug. 30, 2002; 321(5):851-62.
Gessner et al., "The IgG Fc receptor family," Ann. Hematol. Jun. 1998; 76(6):231-48.
Ghetie et al., "FcRn: the MHC class I-related receptor that is more than an IgG transporter," Immunol. Today, 18:592-598 (1997).
Ghetie et al., "Increasing the serum persistence of an IgG fragment by random mutagenesis," Nat. Biotechnol., 15(7):637-40 (1997).
Ghetie et al., "Multiple roles for the major histocompatibility complex class I-related receptor FcRn," Ann. Rev. Immunol. Apr. 2000; 18:739-66.
Glick et al., Molecular Biotechnology: Principles and Applications of Recombinant DNA, 3rd Edition, Chemical Industry Press, Mar. 2005, p. 168 (with English Translation).
Gobburu et al., "Pharmacokinetics/dynamics of 5c8, a monoclonal antibody to CD154 (CD40 ligand) suppression of an immune response in monkeys," J. Pharmacol. Exp. Ther., 286:925-930 (1998).
Goebl et al., "Neonatal Fe Receptor Mediates Internalization of Fc Transfected Human Endothelial Cells," Molecular Biology of the Cell, Dec. 2008, 19(12):5490-5505.
Goode et al., "The glomerular basement membrane charge-selectivity barrier: an oversimplified concept?" Nephrology Dialysis Transplantation. Sep. 1, 1996; 11(9):1714-6.
Graves et al., "Molecular modeling and preclinical evaluation of the humanized NR-LU-13 antibody," Clin. Cancer Res., 5:899-908 (1999).
Greenwood et al., "Structural motifs involved in human IgG antibody effector functions," Eur J Immunol., 23(5):1098-104 (1993).
Gurbaxani et al., "Analysis of a family of antibodies with different half-lives in mice fails to find a correlation between affinity for FcRn and serum half-life," Mol Immunol, Mar. 2006, 43(9):1462-73. Epub Sep. 1, 2005.
Guyre et al., "Increased potency of Fc-receptor-targeted antigens," Cancer Immunol. Immunother. Nov. 1997; 45(3-4):146-8.
Haakenstad et a., "The disappearance kinetics and glomerular deposition of small-latticed soluble immune complexes," Immunology, 47(3):407-14 (1982).
Hamers-Casterman et al., "Naturally occurring antibodies devoid of light chains," Nature, Jun. 3, 1993, 363(6428):446-8.
Hamilton, "Molecular engineering: applications to the clinical laboratory," Clin. Chem., 39(9):1988-97 (1993).
Hanson et al., "Catalytic antibodies and their applications," Curr. Opin. Biotechnol., 16:631-636 (2005).
Haringman et al., "A randomized controlled trial with an anti-CCL2 (anti-monocyte chemotactic protein 1) monoclonal antibody in patients with rheumatoid arthritis," Arthritis Rheum., 54(8):2387-92 (2006).
He et al., "Humanization and pharmacokinetics of a monoclonal antibody with specificity for both E- and P-selectin," J. Immunol., 160:1029-35 (1998).
Hebert LA, "The clearance of immune complexes from the circulation of man and other primates," Am J Kidney Dis., 17(3):352-61 (1991).
Heyman, "Feedback regulation by IgG antibodies," Immunol Lett., 88(2):157-61 (2003).
Hinton et al., "An engineered human IgG1 antibody with longer serum half-life," J. Immunol., 176(1):346-56 (2006).
Hinton et al., "Engineered human IgG antibodies with longer serum half-lives in primates," J. Biol. Chem., 279(8):6213-6 (2004).
Hird et al., "Tumour localisation with a radioactively labelled reshaped human monoclonal antibody," Br J Cancer, Nov. 1991;64(5):911-4.
Hironiwa et al., "Calcium-dependent antigen binding as a novel modality for antibody recycling by endosomal antigen dissociation," MAbs. Jan. 2016; 8(1):65-73. doi: 10.1080/19420862.2015.1110660. Epub Oct. 23, 2015.
Hjelm et al., "Antibody-mediated regulation of the immune response," Scand J Immunol., Sep. 2006;64(3):177-84.
Hong et al., "Enhanced cellular uptake and transport of polyclonal immunoglobulin G and fab after their cationization," J Drug Target., 2000;8(2):67-77.
Hoogenboom, "Selecting and screening recombinant antibody libraries," Nat. Biotechnol., Sep. 1, 2005; 23(9):1105-16.
Horn et al., "Analysis of the binding of pro-urokinase and urokinase-plasminogen activator inhibitor-1 complex to the low density lipoprotein receptor-related protein using a Fab fragment selected from a phage-displayed Fab library," J Biol Chem., May 19, 1995;270(20):11770-5.
Horton et al., "Potent in vitro and in vivo activity of an Fc-engineered anti-CD19 monoclonal antibody against lymphoma and leukemia," Cancer Res., Oct. 1, 2008;68(19):8049-57. doi: 10.1158/0008-5472.CAN-08-2268.
Hotzel et al., "A strategy for risk mitigation of antibodies with fast clearance," mAbs, Nov.-Dec. 2012, 4(6):753-60. doi: 10.4161/mabs.22189.
Hu et al., "Combinatorial libraries against libraries for selecting neoepitope activation-specific antibodies," Proc. Natl. Acad. Sci. USA, Apr. 2010, 107(14):6252-57.
Hwang et al., "Use of human germline genes in a CDR homology-based approach to antibody humanization," Methods, 36:35-42 (2005).
Idusogie et al., "Engineered antibodies with increased activity to recruit complement," J Immunol., Feb. 15, 2001;166(4):2571-5.
Idusogie et al., "Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc," J Immunol., Apr. 15, 2000;164(8):4178-84.
Igawa et al., "Antibody optimization technologies for developing next generation antibody therapeutics," Bio Industry, 28(7):15-21 (2011) (with English translation).
Igawa et al., "Antibody recycling by engineered pH-dependent antigen binding improves the duration of antigen neutralization," Nat Biotechnol., 28(11):1203-7 (2010).
Igawa et al., "Engineered monoclonal antibody with novel antigen-sweeping activity in vivo," PLoS One, 8(5):e63236 (2013).
Igawa et al., "Engineering the variable region of therapeutic IgG antibodies," MAbs. May-Jun. 2011; 3(3):243-52. Epub May 1, 2011.
Igawa et al., "Reduced elimination of IgG antibodies by engineering the variable region," Protein Eng Des Sel., 23(5):385-92 (2010).
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2011/077619, dated Jun. 4, 2013, 8 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2012/058603, dated Oct. 8, 2013, 11 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2012/075083, dated Apr. 1, 2014, 8 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2012/075092, dated Apr. 1, 2014, 10 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2012/081185, dated Jun. 3, 2014, 9 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2013/054461, dated Aug. 26, 2014, 6 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2013/072507, dated Feb. 24, 2015, 6 pages.
International Preliminary Report on Patentability for App. Ser. No. PCT/JP2014/059706, dated Oct. 6, 2015, 10 pages.
International Search Report for App. Ser. No. PCT/JP2011/001888, dated Nov. 2, 2011, 7 pages.
International Search Report for App. Ser. No. PCT/JP2011/077619, dated Feb. 28, 2012, 4 pages.
International Search Report for App. Ser. No. PCT/JP2012/006218, dated Mar. 26, 2013, 11 pages.
International Search Report for App. Ser. No. PCT/JP2012/058603, dated May 29, 2012, 2 pages.
International Search Report for App. Ser. No. PCT/JP2012/075083, dated Oct. 23, 2012, 2 pages.
International Search Report for App. Ser. No. PCT/JP2012/075092, dated Dec. 25, 2012, 4 pages.
International Search Report for App. Ser. No. PCT/JP2012/081185, dated Feb. 26, 2013, 9 pages.
International Search Report for App. Ser. No. PCT/JP2013/054461, dated May 7, 2013, 7 pages.
International Search Report for App. Ser. No. PCT/JP2013/072507, dated Oct. 29, 2013, 4 pages.
International Search Report for App. Ser. No. PCT/JP2014/059706, dated Jul. 15, 2014, 4 pages.
Irani et al., Mol Immunol, Oct. 2015, 67(2 Pt A):171-82. doi: 10. 1016/ j. molimm. 2015. 03. 255. Epub Apr. 18, 2015.
Ishii et al., "FcRn, a critical regulator of antibody pharmacokinetics," Folia Pharmacol. Jpn., 136(5):280-284 (2010) (with English translation).
Ito et al., "The His-probe method: effects of histidine residues introduced into the complementarity-determining regions of antibodies on antigen-antibody interactions at different pH values," FEBS Lett., 309:85-88 (1992).
Jaeger, Clinical Immunology and Allergology, M.: Medicina, 1990, 2:484-5 (with English translation).
Janeway et al., Immunobioiogy, 5th edition. Jun. 1, 2001. Excerpt from Chapter 3.
Janeway et al., Immunobioiogy, 5th edition. Jun. 1, 2001. Excerpt from Chapter 4.
Janeway et al., Immunobiology, 3rd edition 1997, Garland Publishing, Inc., pp. 3:1-3:11. *
Janeway et al., Immunobiology, The Immune System in Health and Disease, 3rd Edition, 1997 Garland Publishing Inc., pp. 3:1-3:11.
Jefferis et al., "Interaction sites on human IgG-Fc for FcgammaR: current Models," Immunol Lett., 82(1-2):57-65 (2002).
Johnson et al., "Cation exchange-HPLC and mass spectrometry reveal C-terminal amidation of an IgG1 heavy chain," Anal. Biochem., 360:75-83 (2007).
Jones et al., "Identification and removal of a promiscuous CD4+ T cell epitope from the C1 domain of factor VIII," Thromb. Haemost., 3:991-1000 (2005).
Junghans et al., "The protection receptor for IgG catabolism is the beta2-microglobulin-containing neonatal intestinal transport receptor," Proc Natl Acad Sci U S A., May 28, 1996;93(11):5512-6.
Juszczak et al., "Ipilimumab: a novel immunomodulating therapy causing autoimmune hypophysitis: a case report and review," Eur J Endocrinol., Jul. 2012; 167(1):1-5. doi: 10.1530/EJE-12-0167. Epub Apr. 10, 2012.
Kamei et al., "Quantitative methods for developing Fc mutants with extended half-lives," Biotechnol Bioeng., Dec. 20, 2005;92(6):748-60.
Kashmiri et al., "Generation, characterization, and in vivo studies of humanized anticarcinoma antibody CC49," Hybridoma, 14:461-473 (1995).
Katayose et al., "MUC1-specific targeting immunotherapy with bispecific antibodies: inhibition of xenografted human bile duct carcinoma growth," Cancer Res. Sep. 15, 1996; 56(18):4205-12.
Khawli et al., "Improved tumor localization and radioimaging with chemically modified monoclonal antibodies," Cancer Biother. Radiopharm. Jun. 1996; 11(3):203-15.
Kim et al., "Antibody Engineering for the Development of Therapeutic Antibodies," Mol. Cells, 20:17-29 (2005).
Kim et al., "Chemical modification to reduce renal uptake of disulfide-bonded variable region fragment of anti-Tac monoclonal antibody labeled with 99mTc," Bioconjug. Chem. May 1999; 10(3):447-53. Online Publication Mar. 20, 1999.
Kim et al., "Lowering of pI by acylation improves the renal uptake of 99mTc-labeled anti-Tac dsFv: effect of different acylating reagents," Nucl. Med. Biol. Nov. 2002; 29(8):795-801.
King, Applications and Engineering of Monoclonal Antibodies, 1998:68-71.
King, Applications and Engineering of Monoclonal Antibodies, Taylor & Francis, ISBN 0-203-21169-3, 2005, pp. 1-236.
Knappik et al., "Fully Synthetic Human Combinatorial Antibody Libraries (HuCAL) Based on Modular Consensus Frameworks and CDRs Randomized with Trinucleotides," J. Mol. Biol., Feb. 11, 2000; 296(1):57-86.
Kobayashi et al., "A monoclonal antibody specific for a distinct region of hen egg-white lysozyme," Mol. Immunol. Apr. 1982; 19(4):619-30.
Kobayashi et al., "The pharmacokinetic characteristics of glycolated humanized anti-Tac Fabs are determined by their isoelectric points," Cancer Res. Jan. 15, 1999; 59(2):422-30.
Kohrt et al., "Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer," J Clin Invest., 122(3):1066-75 (2012).
Komissarov et al., "Site-specific Mutagenesis of a Recombinant Anti-single-stranded DNA Fab Role of Heavy Chain Complementarity-Determining Region 3 Residues in Antigen Interaction," J. Biol. Chem. Oct. 24, 1997; 272(43):26864-70.
Komissarov et al., "Site-specific Mutagenesis of a Recombinant Anti-single-stranded DNA Fab Role of Heavy Chain Complementarity—Determining Region 3 Residues in Antigen Interaction," J. Biol. Chem. Oct. 24, 1997; 272(43):26864-70.
Kuroda et al., "Computer-aided antibody design," Protein Eng Des Sel, Oct. 2012, 25(10):507-21. Epub Jun. 2, 2012.
Kussie et al., "A single engineered amino acid substitution changes antibody fine specificity," J Immunol, Jan. 1, 1994, 152(1):146-52.
Laitinen et al., "Brave new (strept)avidins in biotechnology," Trends Biotechnol., Jun. 2007;25(6):269-77. Epub Apr. 12, 2007.
Lazar et al., "Engineered antibody Fc variants with enhanced effector function," Proc Natl Acad Sci USA, Mar. 14, 2006;103(11):4005-10. Epub Mar. 6, 2006.
Lederman et al., "A Single Amino Acid Substitution in a Common African Allele of the CD4 Molecule Ablates Binding of the Monoclonal Antibody, OKT4," Mol Immunol, Nov. 1991, 28(11):1171-81.
Lee et al., "High-affinity human antibodies from phage-displayed synthetic Fab libraries with a single framework scaffold," J Mol Biol., 340(5):1073-93 (2004).
Leong et al., "Adapting pharmacokinetic properties of a humanized anti-interleukin-8 antibody for therapeutic applications using site-specific pegylation," Cytokine. Nov. 7, 2001; 16(3):106-19.
Lewis et al., "Differential responses of human tumor cell lines to anti-p185HER2 monoclonal antibodies," Cancer Immunol Immunother., 37(4):255-63 (1993).
Li et al., "Apoptotic and antitumor activity of death receptor antibodies require inhibitory Fcγ receptor engagement," Proc Natl Acad Sci U S A., Jul. 3, 2012;109(27):10966-71. doi: 10.1073/pnas.1208698109. Epub Jun. 20, 2012.
Li et al., "CD72 down-modulates BCR-induced signal transduction and diminishes survival in primary mature B lymphocytes," J Immunol., 176(9):5321-8 (2006).
Li et al., "Construction and characterization of a humanized anti-human CD3 monoclonal antibody 12F6 with effective immunoregulation functions," Immunology, Dec. 2005;116(4):487-98.
Li et al., "Inhibitory Fcγ receptor engagement drives adjuvant and anti-tumor activities of agonistic CD40 antibodies," Science, 333(6045):1030-4 (2011).
Li et al., "β-Endorphin omission analogs: Dissociation of immunoreactivity from other biological activities," Proc Natl Acad Sci USA, Jun. 1980, 77(6):3211-4.
Liang et al., "Immunity against a therapeutic xenoprotein/Fc construct delivered by gene transfer is reduced through binding to the inhibitory receptor FcγRIIb," J Gene Med., 13(9):470-7 (2011).
Lin et al., "Preclinical pharmacokinetics, interspecies scaling, and tissue distribution of a humanized monoclonal antibody against vascular endothelial growth factor," J Pharmacol Exp Ther., 288(1):371-8 (1999).
Linder et al., "Design of a pH-dependent cellulose-binding domain," FEBS Lett., Mar. 19, 1999;447(1):13-6.
Liu et al., "Heterogeneity of Monoclonal Antibodies," J. Pharm. Sci. Jul. 2008; 97(7):2426-47. Available Online Sep. 7, 2007.
Lobo et al., "Antibody pharmacokinetics and pharmacodynamics," J. Pharm. Sci., 93:2645-68 (2004).
Lutterbuese et al., "T cell-engaging BiTE antibodies specific for EGFR potently eliminate KRAS- and BRAF-mutated colorectal cancer cells," Proc Natl Acad Sci U S A., Jul. 13, 2010; 107(28):12605-10. doi: 10.1073/pnas.1000976107. Epub Jun. 28, 2010.
Luttrell et al., "Reaction coupling of chelation and antigen binding in the calcium ion-dependent antibody binding of cyclic AMP," J Biol Chem., Nov. 15, 1991;266(32):21626-30.
MacCallum et al., "Antibody-antigen interactions: contact analysis and binding site topography," J. Mol. Biol., 262:732-45 (1996).
Mackay et al., "Selective dysregulation of the FcgammaIIB receptor on memory B cells in SLE," J Exp Med., 203(9):2157-64 (2006).
Maeda et al., "pH-dependent receptor/ligand dissociation as a determining factor for intracellular sorting of ligands for epidermal growth factor receptors in rat hepatocytes," J. Control Release. Jul. 18, 2002; 82(1):71-82.
Maier et al., "Assessment of fully automated antibody homology modeling protocols in molecular operating environment," Proteins. Aug. 2014; 82(8):1599-610. doi: 10.1002/prot.24576. Epub Apr. 23, 2014.
Maini et al., "Double-blind randomized controlled clinical trial of the interleukin-6 receptor antagonist, tocilizumab, in European patients with rheumatoid arthritis who had an incomplete response to methotrexate," Arthritis Rheum., 54:2817-29 (2006).
Malbec et al., "Antibodies against growth factor receptors can inhibit the proliferation of transformed cells via a cis-interaction with inhibitory FcR," Immunol Lett., Mar. 30, 2012;143(1):28-33.
Manger et al., "Fcgamma receptor IIa polymorphism in Caucasian patients with systemic lupus erythematosus: association with clinical symptoms," Arthritis Rheum., 41(7):1181-9 (1998).
Marshall et al., "Rational design and engineering of therapeutic proteins," Drug Discov Today., Mar. 1, 2003;8(5):212-21.
Martin et al., "Crystal structure at 2.8 A of an FcRn/heterodimeric Fc complex: mechanism of pH-dependent binding," Mol. Cell. Apr. 2001; 7(4):867-77.
Martin et al., "Preclinical safety and immune-modulatng effects of therapeutic monoclonal antibodies to interleukin-6 and tumor necrosis factor-α in cynomolgus macaques," J Immunotoxicol., Jul. 1, 2004;1(3):131-9. doi:10.1080/15476910490894904.
Matsumiya et al., "Structural comparison of fucosylated and nonfucosylated Fc fragments of human immunoglobulin G1," J Mol Biol., May 4, 2007;368(3):767-79. Epub Feb. 22, 2007.
Matsunaga et al., "A pH-dependent conformational transition of Abeta peptide and physicochemical properties of the conformers in the glial cell," Biochem. J. Feb. 1, 2002; 361(3):547-56.
Maurer et al., "Antigenicity of polypeptides (poly alpha amino acids): calcium-dependent and independent antibodies," J. Immunol. Sep. 1, 1970; 105(3):567-73.
Maxfield et al., "Endocytic recycling," Nat. Rev. Mol. Cell Biol. Feb. 2004; 5(2):121-32.
Maxwell et al., "Crystal structure of the human leukocyte Fc receptor, FcγRIIa," Nat Struct Biol., May 1999, 6(5):437-42.
Mellman, "The importance of being acid: the role of acidification in intracellular membrane traffic," J Exp Biol, Nov. 1992, 172:39-45.
Merchant et al., "An efficient route to human bispecific IgG," Nat Biotechnol, Jul. 1998, 16:677-681.
Meyer et al., "Bevacizumab immune complexes activate platelets and induce thrombosis in FCGR2A transgenic mice," J Thromb Haemost., 7(1):171-81 (2009). Epub Oct. 30, 2008.
Mi et al., "Targeting the neonatal fc receptor for antigen delivery using engineered fc fragments," J Immunol., 181(11):7550-61 (2008).
Mimoto et al., "Engineered antibody Fc variant with selectively enhanced FcγRIIb binding over both FcγRIIa(R131) and FcγRIIa(H131)," Protein Eng Des Sel., Oct. 2013;26(10):589-98.doi: 10.1093/protein/gzt022. Epub Jun. 5, 2013.
Mohan et al, CALBIOCHEM, "Buffers-A guide for the preparation and use of buffers in biological systems," by Chandra Mohan, Ph.D., CALBIOCHEM Buffers Booklet, 2003, Copyright 2003 EMD Biosciences, Inc., 37 pages.
Mohan et al, CALBIOCHEM, "Buffers—A guide for the preparation and use of buffers in biological systems," by Chandra Mohan, Ph.D., CALBIOCHEM Buffers Booklet, 2003, Copyright 2003 EMD Biosciences, Inc., 37 pages.
Molina et al., "Trastuzumab (Herceptin), a Humanized Anti-HER2 Receptor Monoclonal Antibody, Inhibits Basal and Activated HER2 Ectodomain Cleavage in Breast Cancer Cells," Cancer Res, Jun. 15, 2001, 61(12):4744-9.
Montero-Julian et al., "Pharmacokinetic study of anti-interleukin-6 (IL-6) therapy with monoclonal antibodies: enhancement of IL-6 clearance by cocktails of anti-IL-6 antibodies," Blood, 85(4):917-24 (1995).
Moore et al., "Engineered Fc variant antibodies with enhanced ability to recruit complement and mediate effector functions," MAbs., Mar.-Apr. 2010;2(2):181-9.
Morgan et al., "The N-terminal end of the CH2 domain of chimeric human IgG1 anti-HLA-DR is necessary for C1q, Fc gamma RI and Fc gamma RIII binding," Immunology., 86(2):319-24 (1995).
Muller et al., "VEGF and the Fab fragment of a humanized neutralizing antibody: crystal structure of the complex at 2.4 Å resolution and mutational analysis of the interface," Structure, Sep. 6, 1998; (9):1153-67.
Murata et al., "Anti-Digoxin Fab Variants Generated by Phage Display," Mol. Biotechnol. Jun. 1, 2013:54 (2) :269-77. doi: 10. 1007/s12033-012-9564-1.
Murtaugh et al., "A combinatorial histidine scanning library approach to engineer highly pH-dependent protein switches," Protein Sci., 20(9):1619-31 doi:10.1002/pro 696 (2011).
Muta et al., "A 13-amino-acid motif in the cytoplasmic domain of Fc gamma RIIB modulates B-cell receptor signaling," Nature,368(6466):70-3 (1994).
Nakamura et al., "Fcgamma receptor IIB-deficient mice develop Goodpasture's syndrome upon immunization with type IV collagen: a novel murine model for autoimmune glomerular basement membrane disease," J Exp Med., 191(5):899-906 (2000).
Nam et al., "Current evidence for the management of rheumatoid arthritis with biological disease-modifying antirheumatic drugs: a systematic literature review informing the EULAR recommendations for the management of RA," Ann Rheum Dis., Jun. 2010; 69(6):976-86. doi: 10.1136/ard.2009.126573. Epub May 6, 2010.
Nesterova et al., AACR Abstract No. 656 (2007), Los Angeles, CA. Apr. 4-18, 2007, 3 pages.
Nicholas et al., "Regulation of the immune response. I. Reduction in ability of specific antibody to inhibit long-lasting IgG immunological priming after removal of the Fc fragment," J Exp Med., 129(6):1183-201 (1969).
Niebecker et al., "Safety of therapeutic monoclonal antibodies," Curr Drug Saf., 5(4):275-86 (2010).
Nimmerjahn et al., "Divergent immunoglobulin g subclass activity through selective Fc receptor binding," Science, 310(5753):1510-2 (2005).
Nimmerjahn et al., "Fcgamma receptors as regulators of immune responses," Nat Rev Immunol., 8(1):34-47 (2008).
Nishimoto et al., "Humanized anti-interleukin-6 receptor antibody treatment of multicentric Castleman disease," Blood, 106:2627-32 (2005).
Nishimoto et al., "Interleukin 6: from bench to bedside," Nat. Clin. Pract. Rheumatol., 2:619-626 (2006).
Nishimoto et al., "Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease," Blood, Nov. 15, 2008;112(10):3959-64. doi: 10.1182/blood-2008-05-155846. Epub Sep. 10, 2008.
Non-Final Office Action for U.S. Appl. No. 15/230,904, dated May 25, 2017, 9 pages.
Nordlund et al., "Introduction of histidine residues into avidin subunit interfaces allows pH-dependent regulation of quaternary structure and biotin binding," FEBS Lett., Dec. 18, 2003;555(3):449-54.
Ober et al., "Visualizing the Site and Dynamics of IgG Salvage by the MHC Class I-Related Receptor, FcRn," J. Immunol. Feb. 15, 2004; 172(4):2021-9.
O'Donovan et al., "EGFR and HER-2 Antagonists in Breast Cancer," Anticancer Res, May-Jun. 2007, 27(3A):1285-94.
Official Action dated Oct. 13, 2016, issued for EP Application No. 11714860.1 and submitted as evidence during EP opposition; 3 pages.
Olferiev et al., "The role of activating protein 1 in the transcriptional regulation of the human FCGR2B promoter mediated by the -343 G -> C polymorphism associated with systemic lupus erythematosus," J Biol Chem., 282(3):1738-46 (2007).
Onda et al., "Lowering the Isoelectric Point of the Fv Portion of Recombinant Immunotoxins Leads to Decreased Nonspecific Animal Toxicity without Affecting Antitumor Activity," Cancer Res., 61:5070-77 (2001).
Ono et al., "The humanized anti-HMI.24 antibody effectively kills multiple myeloma cells by human effector cell-mediated cytotoxicity," Mol. Immunol. Apr. 1999; 36(6):387-95.
Osbourn et al., "Generation of a panel of related human scFv antibodies with high affinities for human CEA," Immunotechnology Sep. 1, 1996: 2(3):181-96.
Ozhegov et al., Tolkovyi Slovar Russkogo iazyka: 2004, p. 292 (with an English translation of the relevant passage defining "control").
Pakula et al., "Genetic Analysis of Protein Stability and Function," Annu. Rev. Genet., 23:289-310 (1989).
Palladino et al., "Anti-TNF-alpha therapies: the next generation," Nat Rev Drug Discov., Sep. 2003;2(9):736-46.
Pancook et al., "In Vitro Affinity Maturation of Human IgM Antibodies Reactive with Tumor-Associated Antigens," Hybrid Hybridomics. Oct. 1, 2001: 20(5-6):383-96.
Papista et al., "Dysfunctions of the IgA system: a common link between intestinal and renal diseases," Cell. Mol. Immunol. Mar. 2011: 8(2):126-34. doi:10.1038/cmi.2010.69. Epub Jan. 31, 2011.
Pardridge et al., "Enhanced cellular uptake and in vivo biodistribution of a monoclonal antibody following cationization," J Pharm Sci., Aug. 1995;84(8):943-8.
Pardridge et al., "Enhanced Endocytosis in Cultured Human Breast Carcinoma Cells and In Vivo Biodistribution in Rats of a Humanized Monoclonal Antibody after Cationization of the Protein," J. Pharmacol. Exp. Ther. Jul. 1, 1998; 286(1):548-54.
Patton et al., "An acid dissociation bridging ELISA for detection of antibodies directed against therapeutic proteins in the presence of antigen," J Immunol Methods, Sep. 2005;304(1-2):189-95.
Pavlinkova et al., "Charge-Modified Single Chain Antibody Constructs of Monoclonal Antibody CC49: Generation, Characterization, Pharmacokinetics, and Biodistribution Analysis," Nucl. Med. Biol. Jan. 1999; 26(1):27-34.
Pavlou et al., "The therapeutic antibodies market to 2008," Eur. J. Pharm. Biopharm., 59:389-396 (2005).
Pejchal et al., "A Conformational Switch in Human Immunodeficiency Virus gp41 Revealed by the Structures of Overlapping Epitopes Recognized by Neutralizing Antibodies," J. Virol. Sep. 2009; 83(17):8451-62. doi:10.1128/ JVI. 00685-09. Epub Jun. 10, 2009.
Petkova et al., "Enhanced half-life of genetically engineered human IgG1 antibodies in a humanized FcRn mouse model: potential application in humorally mediated autoimmune disease," Int Immunol., 18(12):1759-69 (2006).
Poduslo et al., "Polyamine modification increases the permeability of proteins at the blood-nerve and blood-brain barriers," J. Neurochem., 66:1599-1609 (1996).
Pons et al., "Energetic analysis of an antigen/antibody interface: Alanine scanning mutagenesis and double mutant cycles on the HyHEL-10/lysozyme interaction," Protein Sci. May 1999; 8(5):958-68.
Popov et al., "The Stoichiometry and Affinity of the Interaction of Murine Fc Fragments with the MHC Class I-Related Receptor, FcRn," Mol Immunol, Apr. 1996, 33(6):521-30.
Presta at el., "Humanization of an Anti-Vascular Endothelial Growth Factor Monoclonal Antibody for the Therapy of Solid Tumors and Other Disorders," Cancer Res, Oct. 15, 1997, 57(20):4593-9.
Presta, "Engineering of therapeutic antibodies to minimize immunogenicity and optimize function," Adv. Drug Deliv. Rev. Aug. 7, 2006; 58(5-6):640-56. Epub May 23, 2006.
Presta, "Molecular engineering and design of therapeutic antibodies," Curr Opin Immunol, Aug. 2008, 20(4):460-70. doi : 10.1016/ j.coi.2008. 06.012.
Prickett et al., "A calcium-dependent antibody for identification and purification of recombinant proteins," Biotechniques, Jun. 1989;7(6):580-9.
Product labelling information for Rituxan (Rituximab), dated Nov. 1997, 2 pages.
Qiao et al., "Dependence of antibody-mediated presentation of antigen on FcRn," Proc Natl Acad Sci USA, 105(27):9337-42 (2008).
Radaev et al., "The role of Fc glycosylation and the binding of peptide inhibitors," J Biol Chem., 276(19):16478-83 (2001).
Radaev et al., "The structure of a human type III Fcgamma receptor in complex with Fc," J Biol Chem., May 11, 2001;276(19):16469-77. Epub Jan. 31, 2001.
Raghavan et al., "Analysis of the pH Dependence of the Neonatal Fc Receptor/Immunoglobulin G Interaction Using Antibody and Receptor Variants," Biochemistry, Nov. 14, 1995, 34(45):14649-5.
Rajpal et al., A general method for greatly improving the affinity of antibodies by using combinatorial libraries, Proc. Natl. Acad. Sci. USA, 102:8466-71 (2005).
Ramos et al., "Evaluation of CA-125 and soluble CD-23 in patients with pelvic endometriosis: a case-control study," Rev. Assoc. Med. Bras. (1992). Jan.-Feb. 2012; 58(1):26-32.
Raposo et al., "Epitope-specific antibody response is controlled by immunoglobulin Vh polymorphisms," J. Exp. Med. Mar. 10, 2014: 211(3):405-11.doi:10.1084/jem.20130968. Epub Feb. 17, 2014.
Rathanaswami et al., "Demonstration of an in vivo generated sub-picomolar affinity fully human monoclonal antibody to interleukin-8," Biochem. Biophys. Res. Commun., 334:1004-13 (2005).
Ravetch et al., "Immune inhibitory receptors," Science, 290(5489):84-9 (2000).
Reddy et al., "Elimination of Fc Receptor-Dependent Effector Functions of a Modified IgG4 Monoclonal Antibody to Human CD4," J. Immunol. Feb. 15, 2000; 164(4):1925-33.
Reichert et al., "Development trends for monoclonal antibody cancer therapeutics," Nat. Rev. Drug Discovery May 2007; 6(5):349-56.
Reichert et al., "Monoclonal antibody successes in the clinic," Nat. Biotechnol., 23:1073-78 (2005).
Reimann et al., "A humanized form of a CD4-specific monoclonal antibody exhibits decreased antigenicity and prolonged plasma half-life in rhesus monkeys while retaining its unique biological and antiviral properties," AIDS Res Hum Retroviruses, Jul. 20, 1997;13(11):933-43.
Restriction Requirement in U.S. Appl. No. 14/402,574, dated Feb. 11, 2016, 10 pages.
Reverberi et al., "Factors affecting the antigen-antibody reaction," Blood Transfus., Nov. 2007;5(4):227-40. doi: 10.2450/2007.0047-07.
Rich et al., "Grading the commercial optical biosensor literature-Class of 2008: 'The Mighty Binders'," J. Mol. Recognit., 23(1):1-64 (2010). doi: 10.1002/jmr.1004.
Rich et al., "Grading the commercial optical biosensor literature—Class of 2008: ‘The Mighty Binders’," J. Mol. Recognit., 23(1):1-64 (2010). doi: 10.1002/jmr.1004.
Richards et al., "Optimization of antibody binding to FcgammaRIIa enhances macrophage phagocytosis of tumor cells," Mol Cancer Ther., 7(8):2517-27 (2008).
Riechelmann et al., "Phase I trial with the CD44v6-targeting immunoconjugate bivatuzumab mertansine in head and neck squamous cell carcinoma," Oral Oncol., Sep. 2008; 44(9):823-9. doi: 10.1016/j.oraloncology.2007.10.009. Epub Jan. 18, 2008.
Robles-Carrillo et al., "Anti-CD40L immune complexes potently activate platelets in vitro and cause thrombosis in FCGR2A transgenic mice," J Immunol., 185(3):1577-83 (2010).
Roitt et al., Immunology, M., Mir, (2000), pp. 110-111 (in Russian, with what is believed to be a published English equivalent of those pages taken from Roitt et al., "Antibody Structure and Function," Immunology, Fifth Ed., (1998), pp. 80-81).
Roitt et al., Immunology, Moscow: Mir, 2000, pp. 373-374 (with English translation).
Rojas et al., "Formation, distribution, and elimination of infliximab and anti-infliximab immune complexes in cynomolgus monkeys," J Pharmacol Exp Ther., May 2005;313(2):578-85. Epub Jan. 12, 2005.
Roopenian et al., "FcRn: the neonatal Fc receptor comes of age," Nat Rev Immunol., 7(9):715-25 (2007).
Rothe et al., "Ribosome display for improved biotherapeutic molecules," Expert Opin. Biol. Ther., 6:177-187 (2006).
Rudge et al.,, "VEGF Trap complex formation measures production rates of VEGF, providing a biomarker for predicting efficacious angiogenic blockade," Proc Natl Acad Sci U S A., 104(47):18363-70 (2007).
Rudikoff et al., "Single amino acid substitution altering antigen-binding specificity," Proc Natl Acad Sci U S A., Mar. 1982;79(6):1979-83.
Rudikoff et al., Proc Natl Acad Sci U S A. Mar. 1982;79(6):1979-83. *
Ryman et al., "Pharmacokinetics of Monoclonal Antibodies," CPT Pharmacometrics Syst Pharmacol, Sep. 2017, 6(9):576-588. doi: 10.1002/psp4.12224. Epub Jul. 29, 2017.
Salfeld et al., "Isotype selection in antibody engineering," Nat. Biotechnol., 25:1369-72 (2007).
Salmon et al., "Fc gamma RIIA alleles are heritable risk factors for lupus nephritis in African Americans," J Clin Invest., 97(5):1348-54 (1996).
Sarkar et al., Rational cytokine design for increased lifetime and enhanced potency using pH-activated "histidine switching," Nat Biotechnol., 20(9):908-13 (2002).
Sato et al., "Reshaping a human antibody to inhibit the interleukin 6-dependent tumor cell growth," Cancer Res., 53:851-856 (1993).
Satoh et al., "Non-fucosylated therapeutic antibodies as next-generation therapeutic antibodies," Expert Opin Biol Ther., 6(11):1161-73 (2006).
Scappaticci et al., "Arterial thromboembolic events in patients with metastatic carcinoma treated with chemotherapy and bevacizumab," J Natl Cancer Inst., 99(16):1232-9 (2007).
Schaeffer et al., "The rat glomerular filtration barrier does not show negative charge selectivity," Microcirculation. Oct. 2002; 9(5):329-42.
Schier et al., "Isolation of Picomolar Affinity Anti-c-erbB-2 Single-chain Fv by Molecular Evolution of the Complementarity Determining Regions in the Center of the Antibody Binding Site," J. Mol. Biol. Nov. 8, 1996: 263(4):551-67.
Schmitz et al., "Phage display: a molecular tool for the generation of antibodies-a review," Placenta Mar. 2000; vol. 21 Suppl A:S106-12.
Schmitz et al., "Phage display: a molecular tool for the generation of antibodies—a review," Placenta Mar. 2000; vol. 21 Suppl A:S106-12.
Schroeder et al., "Similarity and divergence in the development and expression of the mouse and human antibody repertoires," Dev Comp Immunol., 2006;30(1-2):119-35.
Schroter et al., "A generic approach to engineer antibody pH-switches using combinatorial histidine scanning libraries and yeast display," MAbs., 2015;7(1):138-51. doi: 10.4161/19420862.2014.985993.
Schulke et al., "The homodimer of prostate-specific membrane antigen is a functional target for cancer therapy," Proc Natl Acad Sci U S A., Oct. 28, 2003;100(22):12590-5.
Schuster et al., "Signaling of human ciliary neurotrophic factor (CNTF) revisited. The interleukin-6 receptor can serve as an alpha-receptor for CTNF," J Biol Chem., Mar. 14, 2003;278(11):9528-35.
Seda et al., "B-cell receptor signalling and its crosstalk with other pathways in normal and malignant cells," Eur J Haematol., Aug. 1, 2014. doi: 10.1111/ejh.12427.
Sharifi et al., "Improving monoclonal antibody pharmacokinetics via chemical modification," Q J Nucl Med., Dec. 1998;42(4):242-9.
Shields et al., "High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R," J. Biol. Chem., 276:6591-6604 (2001) (Epub Nov. 28, 2000).
Shinkawa et al., "The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity," J Biol Chem., Jan. 31, 2003;278(5):3466-73. Epub Nov. 8, 2002.
Shire et al., "Challenges in the development of high protein concentration formulations," J. Pharm. Sci., 93:1390-1402 (2004).
Sigma-Aldrich, "Product Information: Monoclonal Anti-Flag ® M1, Clone M1 produced in mouse, purified immunoglobulin," Sigma-Aldrich.com, Catalog No. F3040. Retrieved from the Internet on Nov. 5, 2013 at: http://www.sigmaaldrich.com/content/dam/sigma-aldrich/does/Sigma/Datasheet/f3040dat.pdf.
Sims et al., "HMGB1 and RAGE in inflammation and cancer," Annu Rev Immunol., 28:367-88 (2010).
Singer et al., "Genes & Genomes," Moscow, "Mir," 1998;1:63-64.
Singer et al., Genes & Genomes, 1991; pp. 67-69.
Smith et al., "FcgammaRIIB in autoimmunity and infection: evolutionary and therapeutic implications," Nat Rev Immunol., 10(5):328-43 (2010).
Sondermann et al., "Crystal structure of the soluble form of the human Fcγ-receptor IIb: a new member of the immunoglobulin superfamily at 1.7 A resolution," EMBO J Journal, Mar. 1, 1999, 18(5):1095-103.
Sondermann et al., "Molecular Basis for Immune Complex Recognition: A Comparison of Fc-Receptor Structures," J Mol Biol. Jun. 8, 2001;309(3):737-49.
Sondermann et al., "The 3.2-A crystal structure of the human IgG1 Fc fragment-Fc gammaRIII complex," Nature, Jul. 20, 2000;406(6793):267-73.
Stearns et al., "The interaction of a Ca2+-dependent monoclonal antibody with the protein C activation peptide region. Evidence for obligatory Ca2+ binding to both antigen and antibody," J Biol Chem., Jan. 15, 1988;263(2):826-32.
Stepanov, Chapter 4, Primary Structure of Protein, 4.1 Primary structure as a level of protein organization, Molecular biology. Structure and functions of proteins. M.: Nauka, 2005:61-62.
Stewart et al., "Site-directed mutagenesis of a catalytic antibody: an arginine and a histidine residue play key roles," Biochemistry, 33(8):1994-2003 (1994).
Strand et al., "Biologic therapies in rheumatology: lessons learned, future directions," Nat. Rev. Drug Discov., 6:75-92 (2007).
Strohl, Optimization of Fc-mediated effector functions of monoclonal antibodies, Curr Opin Biotechnol., Dec. 2009;20(6):685-91. doi: 10.1016/j.copbio.2009.10.011. Epub Nov. 4, 2009.
Su et al., Expression profile of FcgammaRIIb on leukocytes and its dysregulation in systemic lupus erythematosus, J Immunol., 178(5):3272-80 (2007).
Summary of information about antibodies in Examples of patent (document submitted in EP opposition and posted by EPO on Apr. 13, 2018); 3 pages.
Supplementary data provided by opponent for EP Application No. 11714860.1 (document submitted in EP opposition and posted by EPO on Feb. 20, 2018); 3 pages.
Suzuki et al., "Importance of neonatal FcR in regulating the serum half-life of therapeutic proteins containing the Fc domain of human IgG1: a comparative study of the affinity of monoclonal antibodies and Fc-fusion proteins to human neonatal FcR," J. Immunol., 184(4):1968-76 (2010).
Tabrizi et al., "Elimination mechanisms of therapeutic monoclonal antibodies," Drug Discov Today, 11(1-2):81-8 (2006).
Takeuchi et al., "The Japanese experience with biologic therapies for rheumatoid arthritis," Nat Rev Rheumatol., Nov. 2010; 6(11):644-52. doi: 10.1038/nrrheum.2010.154. Epub Sep. 28, 2010.
Tan et al., "Engineering the isoelectric point of a renal cell carcinoma targeting antibody greatly enhances scFv solubility," Immunotechnology. Oct. 1998; 4(2):107-14.
Tanabe et al., "Characterization of the Monoclonal Antibodies Against Human Protein C Specific for Calcium Ion-induced Conformers," Japanese Journal of Thrombosis and Hemostasis, 1992, 3(1):29-35.
Tanzi et al., "Twenty years of the Alzheimer's disease amyloid hypothesis: a genetic perspective," Cell, Feb. 2005, 120(4):545-55. (doi:10.1016/j.ce11.2005.02.008; PMID 15734686).
Tarditi et al., "Selective high-performance liquid chromatographic purification of bispecific monoclonal antibodies," J. Chromatogr., 599(1-2):13-20 (1992).
Teeling et al., "The Biological Activity of Human CD20 Monoclonal Antibodies Is Linked to Unique Epitopes on CD20," J. Immunol. Jul. 1, 2006; 177(1):362-71.
Ten Kate, et al., "Effect of isoelectric point on biodistribution and inflammation: imaging with indium-111-labelled IgG," Eur. J. Nucl. Med. Jun. 1990; 17(6-8):305-9 (abstract) [Database BIOSIS Accession No. 1991910742 20].
Trinh et al., "Ipilimumab in the treatment of melanoma," Expert Opin Biol Ther., Jun. 2012; 12(6):773-82. doi: 10.1517/14712598.2012.675325. Epub Apr. 14, 2012.
Tsuchiya, Credit Suisse Seminar, "Therapeutic Antibody," at Fuji-Gotemba Laboratories, p. 21, Sep. 22, 2006. English translation.
Tsurushita et al., "Design of humanized antibodies: From anti-Tac to Zenapax," Methods, 36:69-83 (2005).
U.S. Appl. No. 12/295,039, Igawa et al., filed Jan. 20, 2009.
U.S. Appl. No. 12/936,587, Igawa et al., filed Jan. 3, 2011 (abandoned).
U.S. Appl. No. 12/936,587, Igawa et al., filed Jan. 3, 2011.
U.S. Appl. No. 13/595,139, Igawa et al., filed Aug. 27, 2012.
U.S. Appl. No. 13/637,415, Igawa et al., filed Feb. 4, 2013.
U.S. Appl. No. 13/889,484, filed May 8, 2013, Igawa et al.
U.S. Appl. No. 13/889,484, Igawa et al., filed May 8, 2013.
U.S. Appl. No. 13/889,512, filed May 8, 2013, Igawa et al.
U.S. Appl. No. 13/889,512, Igawa et al., filed May 8, 2013.
U.S. Appl. No. 13/990,158, filed May 29, 2013, Igawa et al.
U.S. Appl. No. 13/990,158, Igawa et al., filed Mar. 28, 2014.
U.S. Appl. No. 13/990,158, Igawa et al., filed May 29, 2013.
U.S. Appl. No. 14/007,947, filed Sep. 26, 2013, Igawa et al.
U.S. Appl. No. 14/007,947, Igawa et al., filed Sep. 26, 2013.
U.S. Appl. No. 14/347,034, Igawa et al.
U.S. Appl. No. 14/347,034, Igawa et al., filed Mar. 25, 2014.
U.S. Appl. No. 14/347,321, Igawa et al., filed Mar. 26, 2014.
U.S. Appl. No. 14/361,013, Igawa et al., filed May 28, 2014.
U.S. Appl. No. 14/361,031, Igawa et al., filed May 28, 2014.
U.S. Appl. No. 14/377,556, Igawa et al., filed Aug. 8, 2014.
U.S. Appl. No. 14/377,556, Kuramochi et al., filed Aug. 8, 2014.
U.S. Appl. No. 14/379,825, Igawa et al., filed Aug. 20, 2014.
U.S. Appl. No. 14/404,051, Igawa et al., filed Nov. 26, 2014.
U.S. Appl. No. 14/423,269, Igawa et al., filed Feb. 23, 2015.
U.S. Appl. No. 14/423,269, Katada et al., filed Feb. 23, 2015.
U.S. Appl. No. 14/741,786, Igawa et al., filed Jun. 17, 2015.
U.S. Appl. No. 14/781,069, Igawa et al., filed Sep. 29, 2015.
U.S. Appl. No. 14/781,069, Mimoto et al., filed Sep. 29, 2015.
U.S. Appl. No. 15/050,145, Igawa et al. filed Feb. 22, 2016.
U.S. Appl. No. 15/210,353, filed Jul. 14, 2016, Igawa et al.
U.S. Appl. No. 15/210,353, Igawa et al., filed Jul. 14, 2016.
U.S. Appl. No. 15/210,360, filed Jul. 14, 2016, Igawa et al.
U.S. Appl. No. 15/210,360, Igawa et al., filed Jul. 14, 2016.
U.S. Appl. No. 15/230,904, Igawa et al., filed Aug. 8, 2016.
U.S. Appl. No. 15/495,026, filed Apr. 24, 2017, Igawa et al.
U.S. Appl. No. 15/495,026, Igawa et al., filed Apr. 24, 2017.
U.S. Appl. No. 15/725,692, filed Oct. 5, 2017, Igawa et al.
U.S. Appl. No. 15/725,692, Igawa et al., filed Oct. 5, 2017.
U.S. Appl. No. 15/952,945, filed Apr. 13, 2018, Igawa et al.
U.S. Appl. No. 15/952,945, Igawa et al., filed Apr. 13, 2018.
U.S. Appl. No. 15/952,951, filed Apr. 13, 2018, Igawa et al.
U.S. Appl. No. 15/952,951, Igawa et al., filed Apr. 13, 2018.
U.S. Appl. No. 15/977,757, filed May 11, 2018, Igawa et al.
U.S. Appl. No. 15/977,757, Igawa et al., filed May 11, 2018.
U.S. Appl. No. 15/988,348, filed May 24, 2018, Igawa et al.
U.S. Appl. No. 15/988,348, Igawa et al., filed May 24, 2018.
U.S. Appl. No. 16/028,140, filed Jul. 5, 2018, Igawa et al.
U.S. Appl. No. 16/028,140, Igawa et al., filed Jul. 5, 2018.
U.S. Appl. No. 16/108,897, filed Aug. 22, 2018, Igawa et al.
U.S. Appl. No. 16/108,897, Igawa et al., filed Aug. 22, 2018.
U.S. Appl. No. 61/313,102, filed Mar. 11, 2010, Pons.
USPTO Advisory Action Before the Filing of an Appeal Brief and Notice of Non-Compliant Amendment (37 CFR 1.121) in U.S. Appl. No. 14/404,051, dated Jun. 28, 2018, 4 pages.
USPTO Advisory Action Before the Filing of an Appeal Brief in U.S. Appl. No. 14/404,051, filed Aug. 30, 2018, 3 pages.
USPTO Advisory Action in U.S. Appl. No. 13/889,484, dated Jan. 7, 2016, 3 pages.
USPTO Advisory Action in U.S. Appl. No. 14/402,574, dated Feb. 16, 2017, 3 pages.
USPTO Applicant-Initiated Interview Summary in U.S. Appl. No. 14/402,574, dated May 4, 2018, 27 pages.
USPTO Final Office Action in U.S. Appl. No. 13/595,139, dated Aug. 1, 2013, 8 pages.
USPTO Final Office Action in U.S. Appl. No. 13/595,139, dated Feb. 12, 2016, 12 pages.
USPTO Final Office Action in U.S. Appl. No. 13/595,139, dated May 30, 2017, 23 pages.
USPTO Final Office Action in U.S. Appl. No. 13/889,484, dated Aug. 16, 2017, 14 pages.
USPTO Final Office Action in U.S. Appl. No. 13/889,484, dated Aug. 4, 2015, 12 pages.
USPTO Final Office Action in U.S. Appl. No. 14/347,034, dated Oct. 16, 2015, 5 pages.
USPTO Final Office Action in U.S. Appl. No. 14/347,321 dated Jan. 9, 2017, 60 pages.
USPTO Final Office Action in U.S. Appl. No. 14/347,448, dated Feb. 21, 2017, 45 pages.
USPTO Final Office Action in U.S. Appl. No. 14/347,448, dated Nov. 13, 2018, 29 pages.
USPTO Final Office Action in U.S. Appl. No. 14/361,013, dated Jul. 24, 2017, 43 pages.
USPTO Final Office Action in U.S. Appl. No. 14/402,574, dated Jul. 16, 2018, 10 pages.
USPTO Final Office Action in U.S. Appl. No. 14/402,574, dated Oct. 31, 2016, 16 pages.
USPTO Final Office Action in U.S. Appl. No. 14/404,051, dated Oct. 18, 2017, 15 pages.
USPTO Interview Summary in U.S. Appl. No. 14/347,034, dated Aug. 17, 2015, 3 pages.
USPTO Interview Summary in U.S. Appl. No. 14/347,448, dated Aug. 16, 2017, 4 pages.
USPTO Non-Final Office Action for U.S. Appl. No. 15/230,904, dated Jan. 8, 2018, 16 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 13/595,139, dated Aug. 3, 2015, 13 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 13/595,139, dated Mar. 13, 2015, 12 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 13/595,139, dated Nov. 23, 2016, 11 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 13/595,139, dated Sep. 26, 2018, 32 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 13/889,484, dated Apr. 6, 2015, 12 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 13/889,484, dated Nov. 25, 2016, 12 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/347,034, dated Apr. 16, 2015, 9 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/347,034, dated Jan. 8, 2018, 15 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/347,034, dated May 25, 2017, 16 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/347,321, dated May 2, 2016, 34 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/347,321, dated Nov. 13, 2017, 64 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/347,448, dated Mar. 9, 2018, 32 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/361,013, dated Oct. 28, 2016, 33 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/402,574, dated Jan. 16, 2018, 24 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/404,051, dated Dec. 6, 2016, 22 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/974,350, dated Feb. 1, 2017, 61 pages.
USPTO Non-Final Office Action in U.S. Appl. No. 14/974,488, dated Aug. 16, 2017, 88 pages.
USPTO Notice of Allowance in U.S. Appl. No. 13/595,139, dated Nov. 28, 2017, 17 pages.
USPTO Notice of Allowance in U.S. Appl. No. 13/595,139, dated Oct. 11, 2013, 15 pages.
USPTO Notice of Allowance in U.S. Appl. No. 13/595,139, dated Oct. 3, 2017, 7 pages.
USPTO Notice of Allowance in U.S. Appl. No. 13/889,484, dated Nov. 16, 2017, 18 pages.
USPTO Notice of Allowance in U.S. Appl. No. 14/347,034, dated Feb. 17, 2016, 6 pages.
USPTO Office Action in U.S. Appl. No. 14/347,448, dated May 26, 2016, 54 pages.
USPTO Office Action in U.S. Appl. No. 14/402,574, dated May 6, 2016, 26 pages.
USPTO Reply to Non-Final Office Action in U.S. Appl. No. 13/889,484, dated May 25, 2017, 16 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/347,034, dated Dec. 18, 2014, 9 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/347,321, dated Dec. 17, 2015, 10 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/347,448, dated Dec. 21, 2015, 8 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/347,448, dated Nov. 24, 2017, 13 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/361,013, dated Mar. 16, 2016, 15 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/404,051, dated Apr. 4, 2016, 13 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/974,350, dated Aug. 4, 2016, 8 pages.
USPTO Restriction Requirement in U.S. Appl. No. 14/974,488, dated Jan. 24, 2017, 17 pages.
USPTO Restriction Requirement in U.S. Appl. No. 15/015,287, dated Mar. 13, 2017, 18 pages.
Vaccaro et al., "Divergent activities of an engineered antibody in murine and human systems have implications for therapeutic antibodies," Proc Natl Acad Sci USA, Dec. 5, 2006, 103(49):18709-14. Epub Nov. 20, 2006.
Vaccaro et al., "Engineering the Fc region of immunoglobulin G to modulate in vivo antibody levels," Nat Biotechnol., 23(10):1283-8 (2005).
Vaisitti et al., "Cationization of monoclonal anti bodies: Another step towards the "Magic Bullet"?" J. Biol. Regul. Homeost. Agents Jul.-Dec. 2005; 19(3-4):105-12.
Vajdos et al., "Comprehensive Functional Maps of the Antigen-binding Site of an Anti-ErbB2 Antibody Obtained with Shotgun Scanning Mutagenesis," J. Mol. Biol. Jul. 5, 2002; 320(2):415-28.
Van Walle et al., "Immunogenicity screening in protein drug development," Expert Opin. Biol. Ther. Mar. 2007; 7(3):405-18. Published online Feb. 19, 2007.
Vaughn et al., "Structural basis of pH-dependent antibody binding by the neonatal Fc receptor," Structure, 6(1):63-73 (1998).
Verhoeyen et al., "Construction of a reshaped HMFG1 antibody and comparison of its fine specificity with that of the parent mouse antibody," Immunology, Mar. 1993;78(3):364-70.
Verhoeyen et al., "Monoclonal Antibodies in Clinical Oncology," 1991, Edited by AA Epenetos, Chapter 5, pp. 37-43, Chapman and Hall.
Veri et al., "Monoclonal antibodies capable of discriminating the human inhibitory Fcgamma-receptor IIB (CD32B) from the activating Fcgamma-receptor IIA (CD32A): biochemical, biological and functional characterization," Immunology, 121(3):392-404 (2007).
Veri et al., "Therapeutic control of B cell activation via recruitment of Fcgamma receptor IIb (CD32B) inhibitory function with a novel bispecific antibody scaffold," Arthritis Rheum., 62(7):1933-43 (2010).
Vidarsson et al., "IgG subclasses and allotypes: from structure to effector functions," Front. Immunol. Oct. 20, 2014; 5:520. doi: 10.3389/fimmu.2014.00520.
Waelbroeck et al., "The pH Dependence of Insulin Binding," J Biol Chem, Jul. 25, 1982, 257(14):8284-91.
Wang et al., "HMG-1 as a late mediator of endotoxin lethality in mice," Science, 285(5425):248-51 (1999).
Wang et al., "Monoclonal Antibodies with Identical Fc Sequences Can Bind to FcRn Differentially with Pharmacokinetic Consequences," Drug Metabolism and Disposition, Sep. 2011; 39(9):1469-77. doi: 10.1124/dmd.111.039453. Epub May 24, 2011.
Ward et al., "A calcium-binding monoclonal antibody that recognizes a non-calcium-binding epitope in the short consensus repeat units (SCRs) of complement C1r," Mol Immunol., Jan. 1992;29(1):83-93.
Ward et al., "Evidence to support the cellular mechanism involved in serum IgG homeostasis in humans," Int Immunol, Feb. 2003, 15(2):187-95.
Warmerdam et al., Molecular basis for a polymorphism of human Fc gamma receptor II (CD32), J Exp Med., 172(1):19-25 (1990).
Weiner et al., "Monoclonal antibodies: versatile platforms for cancer immunotherapy," Nat Rev Immunol., May 2010; 10(5):317-27. doi: 10.1038/nri2744.
Welch et al., "Adalimumab (Humira) for the Treatment of Rheumatoid Arthritis" Am Fam Physician, Dec. 15, 2008, 78(12):1406-1408.
Wenink et al., "The inhibitory Fc gamma IIb receptor dampens TLR4-mediated immune responses and is selectively up-regulated on dendritic cells from rheumatoid arthritis patients with quiescent disease," J Immunol., Oct. 1, 2009;183(7):4509-20. doi: 10.4049/jimmunol.0900153. Epub Sep. 4, 2009.
Wernersson et al., "IgG-mediated enhancement of antibody responses is low in Fc receptor gamma chain-deficient mice and increased in Fc gamma RII-deficient mice," J Immunol., 163(2):618-22 (1999).
Wiens et al., "Mutation of a Single Conserved Residue in VH Complementarity-Determining Region 2 Results in a Severe Ig Secretion Defect," J. Immunol. Aug. 15, 2001;167(4):2179-86.
Wiens et al., "Somatic Mutation in VH Complementarity-Determining Region 2 and Framework Region 2," J. Immunol. Aug 1, 1997; 159(3):1293-302.
Wikipedia, "Chaotropic agent" [online], retrieved on Nov. 2, 2015. Retrieved from the Internet at: https://en.wikipedia.org/wiki/Chaotropic_agent. 3 pages.
Wilson et al., "An Fcγ receptor-dependent mechanism drives antibody-mediated target-receptor signaling in cancer cells," Cancer Cell, 19(1):101-13 (2011).
Wojciak et al., "The crystal structure of sphingosine-1-phosphate in complex with a Fab fragment reveals metal bridging of an antibody and its antigen," Proc Natl Acad Sci U S A., 106(42):17717-22 (2009).
Wolf et al., "BiTEs: bispecific antibody constructs with unique anti-tumor activity," Drug Discov. Today. Sep. 15, 2005; 10(18):1237-44.
Written Submissions by Opponent 1 (Alexion Pharmaceuticals, Inc.) in Opposition of EP 2006381 dated Apr. 13, 2018, 19 pages.
Written Submissions by Opponent 2 (Novo Nordisk A/S) in Opposition of EP 2006381 dated Apr. 13, 2018, 14 pages.
Written Submissions by Opponent 3 (name Unknown) in Opposition of EP 2006381 dated Apr. 13, 2018, 16 pages.
Wu et al., "Development of motavizumab, an ultra-potent antibody for the prevention of respiratory syncytial virus infection in the upper and lower respiratory tract," J. Mol. Biol., 368:652-665 (2007).
Wu et al., "Humanization of a murine monoclonal antibody by simultaneous optimization of framework and CDR residues," J. Mol. Biol., Nov. 19, 1999; 294(1):151-62.
Wu et al., "Stepwise in vitro affinity maturation of Vitaxin, an ανvβ33-specific humanized mAb," Proc. Natl. Acad. Sci. USA. May 26, 1998; 95(11):6037-42.
Wu et al., "Ultra-potent Antibodies Against Respiratory Syncytial Virus: Effects of Binding Kinetics and Binding Valence on Viral Neutralization," J Mol Biol, 2005, 350:126-144.
Xiang et al., "Study of B72.3 combining sites by molecular modeling and site-directed mutagenesis," Protein Eng., 13(5):339-44 (2000).
Xiao et al., "Pharmacokinetics of anti-hepcidin monoclonal antibody Ab 12B9m and hepcidin in cynomolgus monkeys," AAPS J., Dec. 2010;12(4):646-57. doi: 10.1208/s12248-010-9222-0. Epub Aug. 25, 2010.
Xolair (omalizumab) Prescribing Information, https://www.gene.com/download/pdf/xolair_prescribing.pdf, Jul. 6, 2016, 27 pages.
Xu et al., "Fc gamma Rs modulate cytotoxicity of anti-Fas antibodies: implications for agonistic antibody-based therapeutics," J Immunol., 171(2):562-8 (2003).
Yamamoto et al., "Molecular studies of pH-dependent ligand interactions with the low-density lipoprotein receptor," Biochemistry, 47(44):11647-52 (2008).
Yamasaki et al., "Pharmacokinetic analysis of in vivo disposition of succinylated proteins targeted to liver nonparenchymal cells via scavenger receptors: importance of molecular size and negative charge density for in vivo recognition by receptors," J. Pharmacol. Exp. Ther., 301:467-477 (2002).
Yang et al., "CDR walking mutagenesis for the affinity maturation of a potent human anti-HIV-1 antibody into the picomolar range," J. Mol. Biol., 254(3):392-403 (1995).
Yang et al., "Dataset of the binding kinetic rate constants of anti-PCSK9 antibodies obtained using the Biacore T100, Protean XPR36, Octet RED384, and IBIS MX96 biosensor platforms," Data Brief, Jul. 27, 2016, 8:1173-83. doi: 10. 1016/ J. dib. 2016.07.044. eCollection Sep. 2016.
Yang et al., "Maximizing in vivo target clearance by design of pH-dependent target binding antibodies with altered affinity to FcRn," mAbs, Oct. 2017, 9(7):1105-1117. doi: 10. 1080/19420862. 2017. 1359455. Epub Aug. 8, 2017.
Yang et al., "Tailoring structure-function and pharmacokinetic properties of single-chain Fv proteins by site-specific PEGylation," Protein Eng. Oct. 1, 2003; 16(10):761-70.
Yarilin, Fundamentals of Immunology, M: Medicina, 1999, pp. 172-174 (with English translation).
Yarilin, Fundamentals of Immunology, M: Medicina, 1999, pp. 175, 182 (with English translation).
Yarilin, Fundamentals of Immunology. M: Medicina, 1999, pp. 169-172, 354-8 (with English translation).
Yarmush et al., "Immunoadsorption: strategies for antigen elution and production of reusable adsorbents," Biotechnol Prog., May-Jun. 1992;8(3):168-78.
Yeung et al., "A Therapeutic Anti-VEGF Antibody with Increased Potency Independent of Pharmacokinetic Half-life," Cancer Res, Apr. 15, 2010, 70(8):3269-77. doi: 10. 1158/ 0008-5472. CAN-09-4580. Epub Mar. 30, 2010.
Yeung et al., "Engineering human IgG1 affinity to human neonatal Fc receptor: impact of affinity improvement on pharmacokinetics in primates," J. Immunol., 182(12):7663-71 (2009).
Yu et al., "Interaction between Bevacizumab and Murine VEGF-A: A Reassessment," Invest Ophthalmol. Vis. Sci. Feb. 1, 2008; 49(2):522-7. doi: 10.1167/iovs.07-1175.
Yuasa et al., "Deletion of fcgamma receptor IIB renders H-2(b) mice susceptible to collagen-induced arthritis," J Exp Med., 189(1):187-94 (1999).
Zalevsky et al., "Enhanced antibody half-life improves in vivo activity," Nat Biotechnol., Feb. 2010;28(2):157-9. doi: 10.1038/nbt.1601. Epub Jan. 17, 2010.
Zalevsky et al., "Enhanced antibody half-life improves in vivo activity," Nat. Biotechnol., 28(2):157-9 (2010).
Zalevsky et al., "The impact of Fc engineering on an anti-CD19 antibody: increased Fcgamma receptor affinity enhances B-cell clearing in nonhuman primates," Blood, 113(16):3735-43 (2009). Epub Dec. 24, 2008.
Zhang et al., "Effective therapy for a murine model of human anaplastic large-cell lymphoma with the anti-CD30 monoclonal antibody, HeFi-1,does not require activating Fc receptors," Blood, 108(2):705-10 (2006).
Zhang et al., "Immune complex/Ig negatively regulate TLR4-triggered inflammatory response in macrophages through Fc gamma RIIb-dependent PGE2 production," J Immunol., Jan. 1, 2009;182(1):554-62.
Zheng et al., "Translational pharmacokinetics and pharmacodynamics of an FcRn-variant anti-CD4 monoclonal antibody from preclinical model to phase I study," Clin Pharmacol Ther., Feb. 2011; 89(2):283-90. doi: 10.1038/clpt.2010.311. Epub Dec. 29, 2010.
Zhou et al., "Interfacial metal and antibody recognition," Proc Natl Acad Sci U S A., 102(41):14575-80 (2005).
Zhu et al., "MHC class I-related neonatal Fc receptor for IgG is functionally expressed in monocytes, intestinal macrophages, and dendritic cells," J. Immunol., 166(5):3266-76 (2001).
Zuckier et al., "Chimeric human-mouse IgG antibodies with shuffled constant region exons demonstrate that multiple domains contribute to in vivo half-life," Cancer Res., 58:3905-08 (1998).
Zwick et al., "The Long Third Complementarity-Determining Region of the Heavy Chain Is Important in the Activity of the Broadly Neutralizing Anti-Human Immunodeficiency Virus Type 1 Antibody 2F5," J. Virol. Mar. 2004; 78(6):3155-61.

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US11371039B2 (en) 2008-04-11 2022-06-28 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US10618965B2 (en) 2011-02-25 2020-04-14 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US11718678B2 (en) 2011-02-25 2023-08-08 Chugai Seiyaku Kabushiki Kaisha Method for altering plasma retention and immunogenicity of antigen-binding molecule
US11827699B2 (en) 2011-09-30 2023-11-28 Chugai Seiyaku Kabushiki Kaisha Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities
US11820793B2 (en) 2011-11-30 2023-11-21 Chugai Seiyaku Kabushiki Kaisha Drug containing carrier into cell for forming immune complex
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11643457B2 (en) 2013-03-13 2023-05-09 Prothena Biosciences Limited Tau immunotherapy
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11912759B2 (en) 2015-01-28 2024-02-27 Novo Nordisk A/S Anti-transthyretin antibodies
US11629185B2 (en) 2015-01-28 2023-04-18 Novo Nordisk A/S Anti-transthyretin antibodies
US11267878B2 (en) 2015-01-28 2022-03-08 Neotope Neuroscience Limited Anti-transthyretin antibodies
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11492393B2 (en) 2016-05-02 2022-11-08 Prothena Biosciences Limited Tau immunotherapy
US11584791B2 (en) 2016-05-02 2023-02-21 Prothena Biosciences Limited Antibodies recognizing tau
US11858980B2 (en) 2016-08-02 2024-01-02 Visterra, Inc. Engineered polypeptides and uses thereof
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11608374B2 (en) 2017-01-30 2023-03-21 Chugai Seiyaku Kabushiki Kaisha Anti-sclerostin antibodies and methods of use
US11958896B2 (en) 2017-05-02 2024-04-16 Prothena Biosciences Limited Antibodies recognizing tau
US11267877B2 (en) 2017-10-06 2022-03-08 Prothena Biosciences Limited Anti-transthyretin antibodies
US11873332B2 (en) 2017-11-29 2024-01-16 Novo Nordisk A/S Lyophilized formulation of a monoclonal antibody against transthyretin
US11926659B2 (en) 2019-03-03 2024-03-12 Prothena Biosciences Limited Antibodies recognizing tau

Also Published As

Publication number Publication date
CN103958547A (zh) 2014-07-30
JP2014528906A (ja) 2014-10-30
TW201817744A (zh) 2018-05-16
RU2018118993A (ru) 2018-11-07
JP2018048120A (ja) 2018-03-29
CN103958547B (zh) 2018-08-03
RU2014117500A (ru) 2015-11-10
JP2019206526A (ja) 2019-12-05
JP6204350B2 (ja) 2017-09-27
CA2850035A1 (en) 2013-04-04
EP2760890A2 (en) 2014-08-06
EP4324850A2 (en) 2024-02-21
BR112014007290A2 (pt) 2017-03-28
SG10201808187TA (en) 2018-10-30
US20240117059A1 (en) 2024-04-11
EP3549956A3 (en) 2020-01-08
JP7001637B2 (ja) 2022-02-03
SG11201401098TA (en) 2014-04-28
JP2022034037A (ja) 2022-03-02
EP2760890B1 (en) 2019-07-24
WO2013046704A2 (en) 2013-04-04
EP3549956A2 (en) 2019-10-09
AU2012313670A1 (en) 2014-03-27
TW201326209A (zh) 2013-07-01
AU2012313670B2 (en) 2017-06-15
KR20140069332A (ko) 2014-06-09
JP6541730B2 (ja) 2019-07-10
AU2017216587B2 (en) 2019-01-31
MX2014003891A (es) 2014-11-26
HK1257946A1 (zh) 2019-11-01
CN108948197A (zh) 2018-12-07
RU2018118993A3 (he) 2019-04-25
AU2017216587A1 (en) 2017-09-07
WO2013046704A3 (en) 2013-05-23
TW201817745A (zh) 2018-05-16
US20140363428A1 (en) 2014-12-11
US20190233525A1 (en) 2019-08-01

Similar Documents

Publication Publication Date Title
US20240117059A1 (en) THERAPEUTIC ANTIGEN-BINDING MOLECULE WITH A FcRn-BINDING DOMAIN THAT PROMOTES ANTIGEN CLEARANCE
US20240158482A1 (en) Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US20170226206A1 (en) Antibodies With Modified Affinity To FcRn That Promote Antigen Clearance
US20230220083A1 (en) Antibodies with modified affinity to fcrn that promote antigen clearance

Legal Events

Date Code Title Description
AS Assignment

Owner name: CHUGAI SEIYAKU KABUSHIKI KAISHA, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:IGAWA, TOMOYUKI;MAEDA, ATSUHIKO;MIMOTO, FUTA;AND OTHERS;SIGNING DATES FROM 20140521 TO 20140526;REEL/FRAME:033136/0434

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4